Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Contents lists available at ScienceDirect
Neuroscience and Biobehavioral Reviews journal homepage: www.elsevier.com/locate/neubiorev
Review
Is there a “metabolic-mood syndrome”? A review of the relationship between obesity and mood disorders Rodrigo B. Mansur a,b,∗ , Elisa Brietzke b , Roger S. McIntyre a a b
Mood Disorders Psychopharmacology Unit (MDPU), University Health Network, University of Toronto, Toronto, Canada Interdisciplinary Laboratory of Clinical Neuroscience (LINC), Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
a r t i c l e
i n f o
Article history: Received 23 April 2014 Received in revised form 19 December 2014 Accepted 31 December 2014 Available online 8 January 2015 Keywords: Mood disorders Major depressive disorder Bipolar disorder Subtypes Obesity Metabolic syndrome Diabetes mellitus Metabolism RDoC Research domain criteria
a b s t r a c t Obesity and mood disorders are highly prevalent and co-morbid. Epidemiological studies have highlighted the public health relevance of this association, insofar as both conditions and its co-occurrence are associated with a staggering illness-associated burden. Accumulating evidence indicates that obesity and mood disorders are intrinsically linked and share a series of clinical, neurobiological, genetic and environmental factors. The relationship of these conditions has been described as convergent and bidirectional; and some authors have attempted to describe a specific subtype of mood disorders characterized by a higher incidence of obesity and metabolic problems. However, the nature of this association remains poorly understood. There are significant inconsistencies in the studies evaluating metabolic and mood disorders; and, as a result, several questions persist about the validity and the generalizability of the findings. An important limitation in this area of research is the noteworthy phenotypic and pathophysiological heterogeneity of metabolic and mood disorders. Although clinically useful, categorical classifications in both conditions have limited heuristic value and its use hinders a more comprehensive understanding of the association between metabolic and mood disorders. A recent trend in psychiatry is to move toward a domain specific approach, wherein psychopathology constructs are agnostic to DSM-defined diagnostic categories and, instead, there is an effort to categorize domains based on pathogenic substrates, as proposed by the National Institute of Mental Health (NIMH) Research Domain Criteria Project (RDoC). Moreover, the substrates subserving psychopathology seems to be unspecific and extend into other medical illnesses that share in common brain consequences, which includes metabolic disorders. Overall, accumulating evidence indicates that there is a consistent association of multiple abnormalities in neuropsychological constructs, as well as correspondent brain abnormalities, with broad-based metabolic dysfunction, suggesting, therefore, that the existence of a “metabolic-mood syndrome” is possible. Nonetheless, empirical evidence is necessary to support and develop this concept. Future research should focus on dimensional constructs and employ integrative, multidisciplinary and multimodal approaches. © 2015 Elsevier Ltd. All rights reserved.
Contents 1. 2.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Is the association of obesity and mood disorders a distinct illness subtype? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Phenomenology, course and treatment outcome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2. Genetic factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3. Environmental risk factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.4. Developmental aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.5. Metabolic systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.6. Brain substrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
∗ Corresponding author at: 399 Bathurst Street, MP 9-325, Toronto, ON, M5T 2S8 Canada. Tel.: +1 416 603 5800; fax: +1 416 603 5368. E-mail address:
[email protected] (R.B. Mansur). http://dx.doi.org/10.1016/j.neubiorev.2014.12.017 0149-7634/© 2015 Elsevier Ltd. All rights reserved.
90 90 90 91 91 91 91 92
90
3.
4.
5.
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
2.7. Animal models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.8. Methodological issues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Obesity and mood disorders are heterogeneous conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Mood disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. Obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . “Metabolic-mood syndrome”: a research agenda . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1. Domains of psychopathology associated to metabolic dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Metabolic dysfunction in mood disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1. Introduction Mood disorders (i.e. major depressive disorder [MDD] and bipolar disorder [BD]) are highly prevalent syndromes. Global prevalence of MDD is estimated to be around 4–5% (Vos et al., 2012; Baxter et al., 2014), whereas BD affects approximately 1.5% of the population (Kessler et al., 2005; Perala et al., 2007). Both conditions often pursue a chronic, unremitting course, which underscores a major impact in morbidity. Mood disorders are leading causes of years lived with disability (YLDs) and disability-adjusted life years (DALY), highlighting MDD and BD as a public health priority (Mathers et al., 2006; Vos et al., 2012; Whiteford et al., 2013). Overweight and obesity have been a major public health concern in the past decades. Epidemiological studies indicate that approximately half of the adult population in Organization for Economic Co-operation and Development (OECD) countries is overweight and about 18% are obese (OECD, 2013). Moreover, this prevalence has increased in all studied countries and in all population groups, regardless of sex, age, race, income or education level (OECD, 2013). Obesity is associated with numerous complications such as type 2 diabetes (T2DM), cardiovascular diseases, musculoskeletal disorders and several forms of cancer, as well as with excess mortality risk (Flegal et al., 2013; Lu et al., 2014; Tobias et al., 2014) and, as a result, has been a cause of growing illness-associated burden and health care costs (Allender and Rayner, 2007; Kelly et al., 2008; Finkelstein et al., 2009, 2010). Obesity and mood disorders are frequently associated. Individuals with MDD have an approximately 50% higher risk of developing obesity, when compared to the general population (Blaine, 2008; Luppino et al., 2010; Toups et al., 2013). The prevalence of obesity is also substantially increased in individuals with BD (Sicras et al., 2008; McIntyre et al., 2010; Kemp et al., 2014). Conversely, overweight and obesity have been reported to increase the risk of onset of significant depressive symptoms and manic episodes (Mather et al., 2009; Luppino et al., 2010; Vogelzangs et al., 2012; Vannucchi et al., 2014). Considering the high impact of obesity and mood disorders in disability and morbidity, the co-occurrence of these conditions is incredibly relevant from a public health perspective and preliminary evidence indicates that a cumulative effect in the associated burden of illness may occur (Everson et al., 2002; Atlantis and Sullivan, 2012; Kemp et al., 2014). Moreover, accumulating evidence has indicated that the association of obesity and mood disorders is not a simple comorbidity. The term comorbidity denotes the co-occurrence of distinct clinical entities. Epidemiological, longitudinal and clinical studies have reported that these illnesses mutually influence each other presentation, trajectory and outcome. Therefore, a bidirectional, convergent relationship would be a more accurate description. Based on these finding some authors have proposed that this cooccurrence constitutes a distinct illness subtype (i.e. “metabolic mood-syndrome”) (McIntyre et al., 2007; Vogelzangs et al., 2011; Levitan et al., 2012). However, many questions about this association remain unanswered. This review aims to discuss the nature
93 93 94 94 94 95 95 96 98 98
of the relationship between obesity and mood disorders and its implications for a subtyping strategy. 2. Is the association of obesity and mood disorders a distinct illness subtype? The concept of a bidirectional relationship between obesity and mood disorders is based not only on the reciprocal predictive risk for each illness, but also on reported differences in clinical manifestations and treatment response linked to this co-occurrence. Furthermore, overlaps of a series of genetic and environmental risk factors, as well as of peripheral and central pathological mechanisms, have been described. 2.1. Phenomenology, course and treatment outcome The presence of obesity has been linked to a distinct and more complicated clinical presentation of mood disorders. In obese or overweight individuals with MDD, when compared to normal weight, there is a significant overrepresentation of atypical features (i.e. mood reactivity, increase in appetite, hypersomnia, leaden paralysis, sensitivity to interpersonal rejection) (Lamers et al., 2010; Cizza et al., 2012; Glaus et al., 2012; Levitan et al., 2012; Toups et al., 2013). In fact, most studies have reported that, in contrast to atypical depression, other specifiers for MDD (e.g. melancholic or undifferentiated depression) have a prevalence of obesity similar to the general population or were more associated to lower weight (Lamers et al., 2010; Cizza et al., 2012; Glaus et al., 2012; Levitan et al., 2012; Chou and Yu, 2013; Lamers et al., 2013; Toups et al., 2013). Due to the cross-sectional nature of the data no conclusions about the causal relationship between obesity and atypical features can be drawn. Although it could be postulated that obesity is a simple consequence of the increase in food intake and lack of exercise associated with atypical depression, results from longitudinal studies indicating that obesity per se is a risk factor for the development of depression challenges this idea. Nonetheless, other phenotypical traits are also associated to higher BMI, including suicide ideation and cognitive symptoms, as well as overall severity of depression (Simon et al., 2008; Marijnissen et al., 2011; Byers et al., 2012). Moreover, obesity/overweight was also associated to a more chronic, persistent trajectory (Vogelzangs et al., 2010; Byers et al., 2012). Evidence from clinical studies in individuals with BD indicates that obesity predisposes to a predominantly depressive illness, insofar the duration of depressive episodes tended to be longer and hospitalization for depression were more frequent (Goldstein et al., 2011). A more severe and chronic course, with higher functional disability, as well as an increased risk of suicide were reported (McIntyre et al., 2008; Calkin et al., 2009). Comorbid anxiety disorders are also more common (Calkin et al., 2009). Furthermore, obese/overweight individuals with BD were shown to have poor cognitive performance, when compared to normal weight patients, with a poor performance in tests measuring attention and
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
psychomotor processing speed (Yim et al., 2012). The presence of depressive symptoms also appears to exert a detrimental effect in obese individuals, as it has been linked to a higher BMI and to the presence of multiple medical comorbidities (Goodman and Whitaker, 2002; Werrij et al., 2006; Zhao et al., 2009). Obesity has also been suggested to negatively impact treatment outcomes in mood disorders. Several studies indicated that higher BMI may be a predictor of poor response to antidepressant in individuals with MDD (Papakostas et al., 2005; Khan et al., 2007; Kloiber et al., 2007; Oskooilar et al., 2009; Uher et al., 2009). In the treatment of BD, obesity was also associated to poor response to pharmacological treatment, including lithium and valproate (Kemp et al., 2010). Conversely, comorbid depression has been reported to be a predictor of unfavorable outcome in different weightloss interventions. Evidence from behavioral, pharmacological and bariatric surgery clinical trials indicates that depressed individuals lose less weight than non-depressed patients and also have poorer long-term weight loss maintenance (Legenbauer et al., 2009, 2011; Ohsiek and Williams, 2011; Pagoto et al., 2013). In fact, individuals that respond well to obesity treatments tend to concurrently display improvements in depressive symptoms and good results have been obtained in trials that employed combined approaches (Simon et al., 2010; Fabricatore et al., 2011; Elder et al., 2012; Busch et al., 2013). 2.2. Genetic factors Mood disorders and obesity are highly heritable illnesses. The heritability of MDD has been estimated between 30% and 50%, whereas for BD is around 50–70% (Sullivan et al., 2000; Smoller and Finn, 2003; Kendler et al., 2006). Obesity also manifests a strong familial influence with the heritability estimates for ranging from 50% to 90% (Stunkard et al., 1986; Hebebrand et al., 2003; Bell et al., 2005). Mood disorders and obesity are thought to follow a polygenic mode of inheritance, with multiple genes contributing to their development (Xia and Grant, 2013; Flint and Kendler, 2014; Kerner, 2014). Genetic studies have found an overlap between genetic risk factors that confer vulnerability for both conditions (Afari et al., 2010; Jokela et al., 2012; Samaan et al., 2013). Interestingly, some of these risk factors were reported to have an interactive effect (Fuemmeler et al., 2009; Rivera et al., 2012; Winham and Biernacka, 2013). For example, the FTO (fat mass and obesity associated) gene has been shown to contribute to obesity, however, a study by Rivera et al. (2012) found that this association was moderated by the presence of depressive symptoms. Conversely, a variant in the TCF7L2 gene, which encodes a transcription factor involved in the Wnt signaling pathway, was associated with protection to BD, but this effect became weaker as BMI increased (Winham and Biernacka, 2013). 2.3. Environmental risk factors Obesity and mood disorders share several risk factors. Traditionally, chronic psychosocial stress is considered one of the most important triggers of mood episodes and has been associated to weight gain and subsequent development of obesity (Altman et al., 2006; Kyrou et al., 2006; Horesh and Iancu, 2010). Inadequate diet and lack of physical exercise are the mainstay of weight gain, but emerging evidence have also highlighted a possible role in MDD and BD onset (Lopresti et al., 2013; Sanhueza et al., 2013; Vancampfort et al., 2013a; Lai et al., 2014). Evidence from epidemiological studies reports a high prevalence of adverse socio-economic situations, including poverty, social isolation, lack of support and low education, in both obese and mood disorders patients (Everson et al., 2002; Sassi et al., 2009; Devaux and Sassi, 2013). Nonetheless, one of the more compelling convergent causative factors is
91
childhood trauma. A history of physical, emotional or sexual abuse is well established as one of the most impactful environmental risk factors for mood disorders (Nanni et al., 2012; Carr et al., 2013; Watson et al., 2013). More recently, accumulating evidence indicates that these traumatic experiences have also a significant impact in metabolic health and increases the risk of obesity, T2DM and metabolic syndrome in adulthood (Gunstad et al., 2006; Midei et al., 2010, 2013; Pervanidou and Chrousos, 2012; Lee et al., 2014a). 2.4. Developmental aspects Converging evidence indicates that mood disorders and obesity also share developmental pathways. Metabolic diseases are influenced by intrauterine and/or early life environment, insofar as exposure to an adverse early environment (e.g. fetal overnutrition or undernutrition) has been consistently associated with increased susceptibilities to obesity, diabetes and cardiovascular disease in adulthood (Moore, 2010; Schulz, 2010; Poston, 2012; Wu et al., 2012). This phenomenon is referred to as developmental origins of health and disease or Barker hypothesis (Bjornsson et al., 2004; Gluckman et al., 2008). Low birth-weight, which is considered a surrogate of adverse intrauterine environment, and early childhood malnutrition have likewise been associated to a higher risk of depression during adolescence and adulthood (Nomura et al., 2007; Galler et al., 2010; Pereira et al., 2012; Wojcik et al., 2013). Moreover, a longitudinal study reported that childhood obesity is an independent risk factor for adult depression (Sanchez-Villegas et al., 2012). Maternal depression during pregnancy has been linked to worse perinatal outcomes, as well as to independently increasing the risk of adolescent depression (Pawlby et al., 2009; Grigoriadis et al., 2013; Pearson et al., 2013; Raisanen et al., 2014), suggesting that it may also be an indicative of a similarly adverse antenatal milieu. In fact, it has been reported that exposure to maternal depressive symptoms in early life affects adiposity, increasing the risk of childhood overweight/obesity, although this may be related to differences in parental style (Adam, 2006; Wang et al., 2013; Ruttle et al., 2014). Among the mechanisms proposed to underlie the long-term effects of adverse early environment are epigenetic processes (e.g. DNA methylations that can cause changes in gene expression). While a comprehensive overview of this topic is outside the scope of this review, it is worth noting that both obesity and depression during pregnancy have been associated with epigenetic modifications (Guenard et al., 2013a,b; Teh et al., 2014). 2.5. Metabolic systems Endocrine and immune-inflammatory regulation have been increasingly more recognized as core features of metabolic dysfunction and have also been shown to be involved in mood disorders. Glucocorticoids and the hypothalamic–pituitary–adrenal (HPA) axis are involved in the physiological response to acute stress. Parts of its effects are to mobilize stored energy and restore energy homeostasis, through actions in the adipose tissue, glucose and insulin regulation, as well as in appetite and feeding. Dysregulation of the HPA axis has been consistently reported in overweight and obese individuals, insofar as studies have shown an association between abdominal adiposity and abnormal activation of the HPA axis, with both higher and lower cortisol responses to stress being reported (Pasquali et al., 2006; Mujica-Parodi et al., 2009; Jones et al., 2012; Kubera et al., 2012; Pasquali, 2012). Conversely, individuals exposed to chronic stress and, as a result, to excessive cortisol were found to have an increase in visceral adipose tissue (Brown et al., 2004). Abnormalities in the HPA axis have also been reliably demonstrated in mood disorders. Individuals with BD display cortisol hypersecretion in euthymia, depression and mania (Cervantes et al., 2001) and show low reactivity to stress, with a
92
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
blunted cortisol response to psychosocial stress (Daban et al., 2005). Individuals with depression, both remitted and symptomatic, also have an increase in cortisol levels (Vreeburg et al., 2009; Stetler and Miller, 2011). Interestingly, studies that compared depressive subtypes have reported that HPA overactivation is related to melancholic features (Wong et al., 2000; Gold and Chrousos, 2002; Stetler and Miller, 2011; Lamers et al., 2013) and not to the atypical symptoms, as it would be expected due to its consistent association with metabolic abnormalities. Accumulating evidence supports the association between mood disorders and immune-inflammatory dysregulation. The presence of higher levels of pro-inflammatory markers (e.g. TNF-a, IL-6, sIL-2R) in the serum of MDD and BD patients was confirmed by multiple meta-analyses (Howren et al., 2009; Dowlati et al., 2010; Munkholm et al., 2013). Inflammatory imbalances were reported in all mood states, including euthymia, as well as in first-episode, drug-naïve and chronic patients. Importantly, as it is well established that adipose tissue is an important source of cytokines, studies that controlled for BMI found much lower, although still significant effect sizes. The role of adiposity as a moderator of immune-inflammatory dysregulation has been highlighted. Obesity is associated to a persistent, low-grade pro-inflammatory state and meta-analytical work has also reported elevated levels of Creactive protein (CRP) in various populations with obesity (Choi et al., 2013). Likewise the HPA axis dysregulation, the differences in inflammatory profile between subtypes of depression has been a focus of study. Individuals with atypical features, when compared to individuals with melancholic features, display higher levels of pro-inflammatory markers, such as CRP, IL-6, TNF-a (Hickman et al., 2013; Lamers et al., 2013). Adipose-derived hormones are another link between mood disorders and metabolic dysfunction. For example, obesity, insulin resistance (IR) and diabetes have been associated with lower levels of adiponectin, an adipose-derived hormone, whereas individuals with MDD and BD were shown to have altered levels of adiponectin (Zeman et al., 2009; Barbosa et al., 2012; Wilhelm et al., 2013). Another member of this family, leptin, is considered to influence obesity, as it exerts a role in the regulation of nutritional intake and energy expenditure. A role in mood disorders has also been proposed for leptin (Lu, 2007; Zupancic and Mahajan, 2011; Barbosa et al., 2012; Milaneschi et al., 2012). Interestingly, a recent study found that the relationship between leptin and depressive symptoms is moderated by central adiposity, as high levels of both leptin and waist circumference incurred in the highest risk of developing depressed mood over a 9-year follow-up (Milaneschi et al., 2014). 2.6. Brain substrates Mood disorders are known to be subserved by alterations in brain structural and functional connectivity. It has been reported that MDD and BD are phenotypically subserved by a disruption of brain networks implicated in emotion regulation, reward processing and cognitive control. Neuroimaging studies have described abnormal integrity in the neural tracts connecting the frontal cortex with the temporal and parietal cortices, as well as with subcortical regions (Lin et al., 2011; Vederine et al., 2011; Liao et al., 2013). Functional connectivity studies have demonstrated decreased connectivity between ventral prefrontal networks and limbic brain structures, including the amygdala, at rest and during performance of tasks that involved emotional processing (Blond et al., 2012; Diener et al., 2012; Strakowski et al., 2012; Vargas et al., 2013). The networks involved are thought to be prominently involved in the modulation of cognition and emotional control (Blond et al., 2012; Diener et al., 2012; Strakowski et al., 2012). More recently, obesity has also been considered to be, at least partially, subserved by abnormal brain networks. Obesity has been
associated with disturbed connectivity in neurocircuits involved in the regulation of motivation and reward (e.g. fronto-occipital, fronto-amygdala networks), both at resting-state and in response to food and non-food rewarding stimuli, indicating that the abnormalities extend beyond appetite regulation (Garcia-Garcia et al., 2012; Kullmann et al., 2012). Data from neuroimaging studies provides evidence for the existence of interactive effects between metabolic disturbances and mood disorders in brain structure and function. In individuals with MDD reductions in subcortical and white matter areas were associated with increased BMI, but not with current depressive symptoms (Cole et al., 2013). In BD, elevated BMI was shown to mediate the decrease of brain whitematter volume and temporal lobe volume (Bond et al., 2011), as well as impaired structural connectivity in temporal, parietal and occipital areas (Kuswanto et al., 2014). Importantly, both studies were done with individuals recently diagnosed with BD, lessening the impact of confounding factors, such as age and cerebrovascular abnormalities. Several neurotransmitters, including, but not limited to, dopamine, opioids and serotonin, are implicated in the brain reward circuitry and the regulation of mood, as well as in the homeostatic regulation of food intake (Cota et al., 2006; Volkow et al., 2011; Russo and Nestler, 2013). Multiple studies reported that dopaminergic neurotransmission in dysregulated in mood disorders. Evidence from pharmacological studies indicates that manipulation of dopamine receptors induce mood alterations (e.g. drugs that increase dopamine levels, such as cocaine, are associated with mood elevation) (D’Aquila et al., 2000). Moreover, medications that target the dopamine receptor (e.g. bupropion, antipsychotics) have been successfully used in the treatment of mood disorders for decades. Imaging studies have also reported abnormalities in the dopaminergic system in both MDD and BD (Chang et al., 2010; Anand et al., 2011; Camardese et al., 2014). In obesity, preclinical and clinical studies have provided evidence of abnormal dopamine signaling, characterized by low dopamine receptor availability, which was proportional to the subject’s BMI, enhanced sensitivity to conditioned stimuli (e.g. high-calorie food), but a decreased sensitivity to rewarding effects (Volkow et al., 2008, 2011; Anand et al., 2011; van de Giessen et al., 2014). An important aspect of brain function that connects mood disorders and metabolic dysfunction is energy metabolism. Abnormalities in cellular bioenergetics, mainly mitochondrial function, have been implicated in the pathophysiology of mood disorders (Clay et al., 2011; Gardner and Boles, 2011; Marazziti et al., 2011; Manji et al., 2012). Neuroimaging studies (i.e., magnetic resonance spectroscopy) provide further support for mitochondrial dysfunction in mood disorders insofar as altered concentrations of brain metabolites including, but not limited to, N-acetyl-aspartate (NAA), lactate and creatine (Cr) have been documented (Iosifescu and Renshaw, 2003; Stork and Renshaw, 2005; Brady et al., 2012; Kraguljac et al., 2012; Chu et al., 2013; Sozeri-Varma et al., 2013; Xu et al., 2013). Conversely, mitochondrial dysfunction has also been implicated in the pathophysiology of obesity (Vernochet and Kahn, 2012; Thrush et al., 2013). Alterations in the brain’s chemical composition, similarly involving NAA and Cr, have also been described (Gazdzinski et al., 2008, 2010; Schmoller et al., 2010). Moreover, the neurotrophin brain-derived neurotrophic factor (BDNF) is considered to be a key mediator of neuroplasticity (Cowansage et al., 2010; Zagrebelsky and Korte, 2014) and is also involved in energy metabolism (Schwartz and Mobbs, 2012; Marosi and Mattson, 2014). This protein has been reported to influence dendritic arborization, axonal growth and synaptic structure and function (Cowansage et al., 2010; Zagrebelsky and Korte, 2014). Allelic variation of the BDNF gene was associated to brain morphology (Ho et al., 2006; Brooks et al., 2014; Forde et al., 2014) and to cognitive function in the general population (Hong et al., 2011;
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Dincheva et al., 2012; Brooks et al., 2014). Decreased peripheral levels of BDNF levels been consistently reported in both MDD and BD in euthymia, depressive and manic episodes (Bocchio-Chiavetto et al., 2010; Fernandes et al., 2011). BDNF is also involved in cellular bioenergetics, modulating neuronal glucose transport and mitochondrial function (Burkhalter et al., 2003; Markham et al., 2012); and in energy homeostasis regulation, as diminished BDNF expression produced hyperphagia and obesity in a mouse model (Unger et al., 2007) and expression of BDNF in the hypothalamus was shown to be inhibited by dietary restriction and enhanced by energy availability (Xu et al., 2003; Unger et al., 2007). Conversely, the frequency of single nucleotide polymorphism (SNP) Val66Met of the BDNF gene was linked to a higher body mass index (BMI) in healthy individuals (Shugart et al., 2009). Finally, the endocannabinoid system (ECS) seems to be involved in the regulation of energy metabolism and may also play a role in the pathophysiology of MDD and BD (Koethe et al., 2007; Hill et al., 2008; Hill and Gorzalka, 2009; Ashton and Moore, 2011). Multiple animal studies have reported antidepressant-like or anxiolytic-like effects of the endocannabinoids (eCBs) (Gobbi et al., 2005; Mitchell and Morris, 2007; Moreira et al., 2008). Altered levels of circulating eCBs in the serum, as well as decreased expression of type 1 cannabinoid receptor (CB1) in the cortex of individuals with mood disorders were reported (Koethe et al., 2007; Hill et al., 2008; Hill and Gorzalka, 2009). More recently, eCB signaling has been considered a critical regulator of the stress response, through modulatory effects in HPA axis activation and behavioral reactions (Hill and McEwen, 2010; Dlugos et al., 2012; Hill and Tasker, 2012). Endogenous eCBs were also reported to be relevant for stress habituation, interacting with different components of the response to chronic stress (Hill et al., 2010). Evidence from animal and human studies indicates that the ECS modulates mitochondrial respiration in neurons and interacts with systems implicated in the regulation of food intake and body weight, including, but not limited to, interacting with energy-related hormones, such as insulin, leptin and ghrelin (Benard et al., 2012; Bermudez-Silva et al., 2012). Dysregulation in the ECS have been described in obesity, insofar as eCB levels were found to be altered in adipose tissue, as well as in the plasma and saliva of obese patients (Bluher et al., 2006; Di Marzo et al., 2009; Bennetzen et al., 2011; Matias et al., 2012). Moreover, several pharmacological studies reported that chronic antagonism of the CB1 receptor significantly reduced body weight, with concomitant improvements of metabolic parameters (Di Marzo and Despres, 2009; Christopoulou and Kiortsis, 2011). A CB1 antagonist, rimonabant, was approved by the European Medicines Agency for the treatment of obesity, however, its commercialization and development was suspended a few years later due to reports of increased anxiety, depression and suicidal ideation (Di Marzo and Despres, 2009; Christopoulou and Kiortsis, 2011). In fact, animal studies have also described that CB1 antagonists impair the antidepressant or antianxiety effects of eCBs, reinforcing the idea that central ECS activity is relevant for mood states, as well as the interconnectedness of molecular pathways mediating both mood and metabolism (Gobbi et al., 2005; Mitchell and Morris, 2007; Moreira et al., 2008). More recently, research in the ECS as a therapeutic target for obesity has focused on the development of neutral antagonists and peripherally restricted antagonists, as well as molecules that indirectly target eCBs levels (e.g. n-3 polyunsaturated fatty acids), which would still have the potential to ameliorate metabolic parameters, but without the reported negative effects on mood (Le Foll et al., 2009; Silvestri and Di Marzo, 2012; Boon et al., 2014). 2.7. Animal models Preclinical studies have provided further evidence of the interconnectedness of mood disorders and obesity. Chronic stress
93
paradigms have been widely used to mimic depression and/or depressive-like behavior in animals (e.g. social avoidance, anhedonia) (Strekalova et al., 2011). In addition to its behavioral effects, chronic stress models have also been shown to impact metabolic processes, being associated to an increase in adiposity, induction of insulin resistance and disruption of lipid regulation (Chuang et al., 2010; Patterson et al., 2013; Lin et al., 2014). Animals fed a high-fat diet (HFD), which readily gain weight, are a model amply used to study human obesity and metabolic syndrome (Buettner et al., 2007). Evidence indicates that HFD mice exhibit a depressivelike phenotype as well, similarly to the observed in stress-based paradigms (Sharma and Fulton, 2013; Hu et al., 2014). Notably, effects of HFD include neurobiological alterations related to depressive phenotypes, such as abnormalities in limbic system activation and reward circuitry, decrease in serotonin and dopamine levels and enhanced HPA axis reactivity (Kim et al., 2013; Sharma and Fulton, 2013; Hu et al., 2014). An animal model of T2DM (i.e. db/db mice, characterized by a mutation in the gene encoding the long isoform of the leptin receptor) was also reported to display behavioral depression (Sharma et al., 2010). Moreover, a study using the Flinders Sensitive Line (FSL) rat, a validated genetic animal model of depression (Overstreet et al., 2005), found that HFD exacerbated depressive-like features in the FSL rats, but did not affect non-depressed rats, suggesting that a more vulnerable rat strain was more susceptible to the detrimental effects of HFD (Abildgaard et al., 2011).
2.8. Methodological issues Notwithstanding the accumulating body of evidence linking obesity and mood disorders, there are important methodological limitations that should be considered. A significant number of studies failed to find an association between these two conditions or could not support the idea that this association incurred in differences in phenomenology, trajectory or treatment response (Papakostas et al., 2005; Dave et al., 2011; Chang and Yen, 2012; Dong et al., 2013; Goldstein et al., 2013; Toups et al., 2013). Inconsistencies in the results have also been frequent in studies evaluating metabolic processes and mood disorders. For example, some studies did not observe alterations in inflammatory markers in MDD (Haack et al., 1999; Steptoe et al., 2003) and most authors agree that not all individuals with depression have a pro-inflammatory profile (Raison et al., 2013). Discrepancies can also be found in the data obtained from animal models, as a study using a chronic social defeat model observed that the depressive features were in fact associated with anorexic behavior (Iio et al., 2012). A separate study documented that rats exposed to mild food restriction had a higher frequency of depressive-like behavior when compared to controls (Iio et al., 2014). This variability in results is meaningful and could be explained by some factors. First, most of the studies did not considered age and gender in the analyses. As it is known that weight and mood disorders, as well as other important clinical characteristics, are distributed differently between age groups or gender (Baskaran et al., 2014), this omission could have biased some of the results. Second, almost all studies used categorical BMI to contrast obesity and normal weight. Although BMI categories are the standard criteria, recent research have highlighted that obesity is characterized by a considerable clinical and pathophysiological heterogeneity. Moreover, it can also be argued that mechanistic studies of a general group of individuals diagnosed with MDD or BD is inherently limited, due to phenotypical and etiopathological heterogeneity of mood disorders that is inextricably linked to its marked interindividual variability in clinical presentation and treatment response. As a result, the identification of subgroups with a more homogenous set of characteristics emerges as a more
94
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
promising strategy to advance our understanding of obesity and mood disorders. 3. Obesity and mood disorders are heterogeneous conditions Obesity and mood disorders are defined by their phenotypic expressions (i.e. accumulation of adipose tissue; altered mood and/or energy levels). Nonetheless, it is well known that the underlying mechanisms leading to the emergence of these symptoms are extremely varied and include several different genetic, behavioral, and environmental factors. Recently, an emerging trend in medical sciences is the subtyping of heterogeneous diseases based on pathophysiological mechanisms (Costa e Silva, 2013; Ozomaro et al., 2013; Yang et al., 2013). This approach has been successfully applied in cancer research, whereas it has been demonstrated that risk factors and treatment response may vary by molecular subtypes (Gonzalez de Castro et al., 2013). For example, a study found that patients with mutated-PIK3CA colorectal cancer benefited from taking aspirin, whereas patients with wild-type PIK3CA cancer did not (Liao et al., 2012). The complex nature of obesity and mood disorders, as well as the challenges in developing effective strategies for prevention and treatment makes them excellent candidates for this approach. 3.1. Mood disorders Mood disorders have a rich history of subtype’s descriptions. Several subgroups have been proposed based on specific combinations of symptoms (i.e. melancholic depression, psychotic depression), onset (seasonal affective disorder, postpartum), etiology (exogenous, endogenous) or severity (Harald and Gordon, 2012). However, the clinical applications of these distinctions were mostly unsuccessful and they were gradually abandoned (van Loo et al., 2012). Currently, two main groups of mood disorders are recognized, depressive disorders and bipolar disorders, with different subcategories within each group (e.g. BD I and II, dysthymia, cyclothymia, substance-induced mood disorders). The diagnostic criteria of mood disorders are based on the combination of different symptoms. A debate about the validity of these diagnostic categories has occurred for decades in psychiatry. Most authors agree that these categories have contributed greatly to the reliability of psychiatric diagnoses and are generally useful in clinical practice to inform treatment planning and prognosis. Nonetheless, they are based solely on phenomenology and do not incorporate developmental and etiopathological features that were recently highlighted by neuroscience research. Genetic and psychometric studies have reported that these symptomatic criteria do not reflect a single underlying factor (Kendler et al., 2013). As a result, two patients can exhibit entirely non-overlapping clusters of symptoms, but receive the same official diagnosis and an identical treatment. Mood disorders are syndromes defined by the presence of an abnormal mood state, either high (mania/hypomania) or low (depression), accompanied by other behavioral, cognitive and neurovegetative alterations. A myriad of pathophysiological processes have been thought to underlie these phenotypical expressions. Currently, it has been hypothesized that the proximal etiological factor is an abnormal structural and functional integrity of neural circuits in distributed networks implicated in cognition and emotional processing (Diener et al., 2012; Strakowski et al., 2012; Phillips and Swartz, 2014). The abnormal brain connectivity is believed to be a consequence of a multifaceted interaction of several genetic and environmental factors (e.g. childhood trauma, psychosocial stress and drug abuse), as well as to involve
different pathophysiological mechanisms that include, but are not limited to, dysfunctions in neurotrophic, immuno-inflammatory and oxidative stress pathways (Strakowski et al., 2012; Moylan et al., 2013; Pfaffenseller et al., 2013). Considering the complex etiopathogenesis and clinical presentation of mood disorders, it is not surprising that treatment outcomes are largely heterogeneous and a significant percentage of the patients do not respond to any of the available treatment options (Perlis et al., 2006; Nierenberg et al., 2010; Bowden et al., 2012). To address this issue, recent clinical studies have attempted to identify more homogeneous and possibly responsive subgroups. For example, the study by Raison et al. (2013) reported that a TNF antagonist (infliximab) did not have a generalized efficacy in treatment-resistant depression. However, when only a subset of patients with high baseline inflammatory biomarkers (i.e. high levels of high-sensitive C-reactive protein) was considered, the experimental treatment significantly improved depressive symptoms, indicating that the subgrouping of individuals and a biomarker informed treatment selection are promising strategies for further research (Raison et al., 2013).
3.2. Obesity Obesity is characterized by excessive accumulation of body fat. Implied in the concept is the notion of a pathological process that will, eventually, culminate in health problems and reduced life expectancy. The World Health Organization (WHO) establishes that a body-mass index (BMI) of 25 kg/m2 or higher is abnormal, wherein a BMI between 25 and 30 kg/m2 is categorized as overweight; a BMI of 30 kg/m2 or more is classified as obese (although there are ethnic differences). Generally, most cases of obesity are thought to be caused by a combination of excessive food consumption and a sedentary lifestyle. However, there is a remarkable variability in how much these factors contribute to cause obesity in different individuals (Despres, 2001; Naukkarinen et al., 2012; Field et al., 2013). Several biological pathways and environmental stimuli are known to be involved in the regulation of body weight, appetite and level of physical activity. Genetic studies have reported specific genes associated to higher adiposity, food intake and response to physical exercise (Ahmad et al., 2013; Fesinmeyer et al., 2013; Tanaka et al., 2013). Hormonal mechanisms for appetite and satiety, as well as energy expenditure regulation are well described (Hussain and Bloom, 2013). The insulin pathway and the development of insulin resistance have been studied for decades (Kahn et al., 2006). Moreover, the adipose tissue, once considered as only a reservoir for lipids, is now recognized as an active endocrine organ that secretes a variety of hormones, including leptin, adiponectin, and resistin, which are involved in energy metabolism (Jackson and Ahima, 2006). The role of the central nervous system (CNS) in obesity has been increasingly recognized (Hussain and Bloom, 2013; Karatsoreos et al., 2013; Jauch-Chara and Oltmanns, 2014). More recently, brain regions and networks implicated in satiety signals, reward to food and addictive behavior; neurohormonal regulatory processes and the effect of mood and psychosocial stress have been described and are currently a focus of research (Hussain and Bloom, 2013; Karatsoreos et al., 2013; Jauch-Chara and Oltmanns, 2014). Taken together, the current evidence supports a disease model of obesity based on the interaction of multiple biological and environmental processes. The existence of a several distinct (albeit interconnected) pathological processes suggests that in different individuals abnormal body weight can be underlied by different mechanisms. This concept is supported by the results of multiple clinical trials for the treatment of obesity, including pharmacological and dietary/behavioral interventions, which showed, at best, small effect sizes and/or important inconsistencies in the results (Gloy et al., 2013; Swift et al., 2014; Yanovski and Yanovski, 2014).
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Another aspect of obesity that has a significant interindividual variability is the presence of metabolic comorbidities, such as T2DM, dyslipidemia, hypertension and cardiovascular diseases. Epidemiological studies have reported a subset of obese individuals that do not present these obesity-related metabolic disturbances (Ferrannini et al., 1997; Karelis et al., 2005; Stefan et al., 2008; Wildman et al., 2008; Primeau et al., 2011). Interestingly, this subgroup, which has been known as “metabolically healthy obese” (MHO), appears to exhibit a different illness trajectory, with a cardiovascular risk and incidence of complications more similar to normal weight individuals (Ferrannini et al., 1997; Iacobellis et al., 2005; Karelis et al., 2005; Primeau et al., 2011; Kramer et al., 2013; Morkedal et al., 2014). Some criteria have been proposed to discriminate healthy from at-risk obese subjects. These criteria include differences in body fat composition/distribution, insulin sensitivity and pro-inflammatory markers; however there is no universally accepted definition of MHO to this date (Primeau et al., 2011; Denis and Obin, 2013). It has also been reported that normal weight individuals may also display obesity associated disorders, which was termed normal-weight obesity (NWO), indicating that BMI is not perfectly correlated to metabolic health (Marques-Vidal et al., 2008; Oliveros et al., 2014). Moreover, the connection between higher BMI with increased mortality rates has recently been challenged. Accumulating evidence has shown that in specific populations, such as individuals with T2DM, chronic kidney disease or heart failure, the presence of overweight may be protective, as this group was reported to have lower mortality when compared to normal weight individuals (Oreopoulos et al., 2008; Jialin et al., 2012; Padwal et al., 2013; Thomas et al., 2014). This phenomenon is known as “obesity paradox” and its significance has been highlighted by findings that in the general population BMIs between 25 and 30 kg/m2 were also predictive of lower mortality (Flegal et al., 2013). Although there are controversies regarding the validity of “obesity paradox” in the general population (Tobias et al., 2014), these results, as well as the existence of the intermediate phenotypes MHO and NWO, suggests that metabolic dysfunction is better conceptualized as a multidimensional construct and it is not just a reflection of weight status. 4. “Metabolic-mood syndrome”: a research agenda It has been proposed that the association between obesity and mood disorders characterizes, in fact, a distinct subtype, the “metabolic-mood syndrome”, whereas alterations in mood and metabolism are clinically connected and mutually influence each other (McIntyre et al., 2007; Vogelzangs et al., 2011; Levitan et al., 2012). However, the methodological issues mentioned above are important limitations and preclude an accurate and definitive description of a subpopulation’s characteristics. Considering the marked heterogeneity of obesity and mood disorders research in the area would greatly benefit from a domain-specific and dimensional approach to the convergent psychopathological and metabolic manifestations. 4.1. Domains of psychopathology associated to metabolic dysfunction Overall, most studies have indicated that individuals with the “metabolic-mood syndrome” have a predominantly depressive illness, with an overrepresentation of atypical features, anxiety and a chronic course. Despite the usefulness of employing clusters of symptoms to define psychiatric disorders, decades of research have shown that this is a limited strategy, as a single symptom can be subserved by disparate pathophysiological process. Within this context, the use of domains of functioning and their constituent
95
dimensional constructs, as proposed by the National Institute of Mental Health (NIMH) Research Domain Criteria Project (RDoC) emerges as a promising strategy for research (Morris and Cuthbert, 2012; Insel, 2014). For example, anhedonia, a core feature of mood disorders, has been recently studied as dysfunction of reward, as defined by responses to emotional stimuli such as food, sex and social interaction (Dillon et al., 2014). Some aspects of the brain’s reward circuitry are relatively well characterized, including the neural systems involved in reward anticipation, consummation and learning, as well as its molecular aspects, particularly the regulation of dopamine transmission (Russo and Nestler, 2013). The focus on a core psychological function and the use of an integrated approach, involving behavioral, neural circuit, and molecular levels of anhedonia, allows a better understanding of this phenomena and how it relates to mood disorders course and treatment. The interaction of mood and metabolism would benefit from such approach. In keeping with the example of anhedonia, reward circuits are also known to be prominently involved in eating behavior and have been a focus of obesity research (Petrovich et al., 2005; Baldo and Kelley, 2007; Volkow et al., 2008; Jauch-Chara and Oltmanns, 2014). This raises several interesting questions, including if and how anhedonia moderates the “metabolic-mood syndrome”? Preliminary evidence comes from the study of Keranen et al. (2010), which evaluated the relationship between anhedonia and eating behavior. Anhedonia, regardless of whether the participant had been diagnosed with a mood disorder, was significantly associated with increased food intake, mainly due to higher rates of uncontrolled, emotional and binge eating (Keranen et al., 2010). Other neuropsychological constructs were also reported to be moderated by the co-occurrence of metabolic and mood disorders. Higher rates of anxiety in individuals with both conditions indicate that alterations in the response to stress may also be involved in this association. Impairment in the psychosocial stress response has been recognized as a core feature of mood disorders clinical expression. Stress is a well-established trigger of mood episodes and stressful life events are one of the strongest predictors of illness onset and relapse (Altman et al., 2006; Horesh and Iancu, 2010; Horesh et al., 2011). Chronic stress exposure is associated with a higher number of mood episodes, as well as with greater symptom severity and persistence (Kim et al., 2007). Stress response and habituation also have an important role in appetite and body weight regulation. Acute stress was shown to increase food intake, especially carbohydrates and/or high caloric meals (Oliver et al., 2000; Hitze et al., 2010; Tryon et al., 2013). The consumption of high-energy foods, in its turn, was reported to attenuate the stress response and its negative effects, including an improvement in mood (Bell et al., 2002; Canetti et al., 2002; Pecoraro et al., 2004; Hitze et al., 2010). This effect appears to be modulated by several features, including gender (Oliver et al., 2000) and history of chronic stress (Tryon et al., 2013), insofar as chronically stressed individuals were reported to have an even greater increase in poststress food craving and caloric consumption, as well as a heightened stress-induced negative mood (Tryon et al., 2013). Consequentially, studies have consistently described that exposure to chronic stress is positively correlated to BMI and fat mass (Tamashiro et al., 2011; Pervanidou and Chrousos, 2012; Tryon et al., 2013). Stress response and adaptation is largely related to a myriad of psychological factors and biological pathways, nonetheless, evidence indicates that eating behavior and body composition are important aspects of it. Therefore, it could be hypothesized that individuals with the “metabolic-mood syndrome” would be more likely to have a history of chronic stress (e.g. childhood trauma, which is a shared risk factor for mood and metabolic disorders) and would respond to acute stress with an even more marked increase in food consumption. However, no study has directly tested this hypothesis to this date.
96
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Evidence indicates that neurocognitive dysfunction is more prominent in overweight/obese individuals with a mood disorder (Yim et al., 2012). Cognitive dysfunction has been consistently demonstrated in MDD and BD and is considered a core feature of mood disorders (Mann-Wrobel et al., 2011; Lee et al., 2012; Bourne et al., 2013; Snyder, 2013). Generalized impairments in several cognitive measures were reported, including, but not limited to, attention, memory, processing speed and executive function (Mann-Wrobel et al., 2011; Lee et al., 2012; Bourne et al., 2013; Snyder, 2013). Several moderators are recognized, such as sociodemographic and clinical features; nonetheless, evidence indicates that neurocognitive impairment is independently associated to mood disorders (Bourne et al., 2013; Snyder, 2013). Conversely, overweight/obesity and metabolic syndrome have been shown to negatively impact a variety of cognitive domains (Gunstad et al., 2007; Taylor and MacQueen, 2007; Gunstad et al., 2010). One of the more intriguing finding from animal studies has been the robust detrimental effect of HFD on cognitive performance (Stranahan et al., 2008; Gault et al., 2010; Porter et al., 2010), as well as the reversibility of the deficits by dietary changes, physical exercise or pharmacological manipulations (Gault et al., 2010; Porter et al., 2010; Pintana et al., 2013; Pipatpiboon et al., 2013; Woo et al., 2013). In humans, impairments in memory, learning executive function and psychomotor speed have also been consistently reported in T2DM (Strachan et al., 1997; Gold et al., 2007; McCrimmon et al., 2012). The relevance of the effects of metabolic diseases in neurocognitive function is underscored by evidence suggesting that weight loss in overweight and obese individuals significantly improves cognitive performance (Siervo et al., 2011). Furthermore, both metabolic and mood disorders have been considered as independent risk factors for the development of late-life mild cognitive impairment and Alzheimer’s dementia (Whitmer et al., 2005; Ownby et al., 2006; Xu et al., 2011; Cheng et al., 2012). Overall, evidence indicates that the role of cognition on the “metabolic-mood syndrome” can be understood in two complementary ways. As metabolic and mood disorders are independently associated to poor cognitive performance, the cooccurrence of these conditions may have a synergistic detrimental effect (Yim et al., 2012), thus, greater cognitive impairment would be consequential. Nonetheless, cognitive dysfunction has also been considered as a vulnerability factor for mood disorders (Correll et al., 2007; Skjelstad et al., 2010). Interestingly, cognitive factors, especially those related to cognitive control, regulation of motivation and reward, as well as the cognitive response to stress, have also been reported to be prominently involved in the development of obesity (Baldo and Kelley, 2007; Mujica-Parodi et al., 2009; Jauch-Chara and Oltmanns, 2014). In that case, neurocognition could be thought to have a more broad modulatory effect in the link between metabolic and mood disorders, concurrently increasing the risk for both conditions and moderating how they affect each other. In conclusion, accumulating evidence indicates that the clinical presentation of the “metabolic-mood syndrome” involves more than just the co-occurrence of obesity and mood symptoms. The association of metabolic and mood disorders seems to broadly impact multiple neuropsychological domains. Preliminary evidence suggests that these effects may be related to several constructs proposed by the RDoC project, including Negative Valence, Positive Valence, Arousal/Regulatory Systems and Cognitive Systems (Morris and Cuthbert, 2012). Further studies, using multimodal and integrative approaches, are still necessary to understand better the multidimensional aspects of the interaction between mood and metabolism.
4.2. Metabolic dysfunction in mood disorders Obesity has been shown to be only one of the several different determinants of metabolic health. Longitudinal studies using the MHO concept have described that metabolic dysfunction, as understood by the risk of obesity-associated conditions (e.g. CVD, stroke and cancer), seems to be also related to the presence of metabolic comorbidities (e.g. insulin resistance, dyslipidemia, hypertension) and to hormonal and inflammatory profiles. Interestingly, a study by Hamer et al. (2012) found that MHO was not associated to a higher risk for depressive symptoms after a 2 year follow-up, whereas metabolically unhealthy individuals (defined by the presence of 2 or more abnormalities in the following: blood pressure, cholesterol, triglycerides, glycated hemoglobin and Creactive protein) did have an elevated risk. Another study reported an intermediate risk of depression in MHO when compared to non-obese and metabolically unhealthy individuals (Jokela et al., 2013). Taken together these studies indicate that the association between obesity and mood symptoms also appears to be dependent on metabolic profile. As discussed above, there are no established criteria for the characterization of MHO. There is also no consensus about how to define NWO and, therefore, there is no single definition of metabolic dysfunction. Most authors agree that metabolic risk is associated to a combination of different clinical factors, including body adiposity (i.e. central obesity, high visceral white adipose tissue), dyslipidemia and hypertension. The co-occurrence of these features defines metabolic syndrome (MS), a diagnosis that was created to assess and predict CVD and T2DM risks. Accumulating evidence indicates that there is also an association between mood disorders and MS, as well as its subcomponents. Individuals with MDD and BD have been shown to have, when compared to the general population, increased waist circumference, higher proportion of visceral adiposity, raised levels of serum lipids and higher incidence of hypertension, as well as, consequently, to meet criteria for MS more frequently (Fagiolini et al., 2005; Veen et al., 2009; Ludescher et al., 2011; Czepielewski et al., 2013; Vancampfort et al., 2013b). However, there is less evidence regarding the interaction of MS with mood disorders when compared to obesity. A meta-analysis of cross-sectional and cohort studies observed a bidirectional association (Pan et al., 2012), nonetheless, the effects of MS on the phenomenology, course and treatment response of mood disorders are mostly unknown. A bidirectional relationship also seems to be case with diabetes mellitus. The rate of mood disorders is increased in T2DM and vice versa, independently of the presence of overweight or obesity (Ali et al., 2006; Barnard et al., 2006). The risk of developing T2DM is increased by approximately 60% in individuals with MDD (Rotella and Mannucci, 2013a) and individuals with diabetes display a significantly elevated risk for developing MDD, independently of other risk factors (e.g., alcohol or drug use, smoking, medical comorbidities and physical limitations) (Rotella and Mannucci, 2013b). Follow-up studies have also reported the presence of altered glucose metabolism prior to the onset of depression (Golden et al., 2008; Hamer et al., 2011). In addition, depressive symptoms are associated with a more severe course of diabetes, including worse glycemic control, a higher incidence of complications and an increased risk of mortality (Lustman et al., 2000; de Groot et al., 2001; Bruce et al., 2013). Conversely, studies have shown increased morbidity in patients with mood disorders and co-morbid T2DM (Ruzickova et al., 2003; Le et al., 2011), whereas, for example, individuals with BD and T2DM were more likely to have a chronic course, rapid cycling and a worse functional impairment, when compared to non-T2DM subjects (Ruzickova et al., 2003). Insulin resistance (IR), which is considered to be a precursor of T2DM, has been understood as a key marker of metabolic
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
97
Fig. 1. Multidimensional aspects of the association between metabolic and neuropsychological phenotypes. Abnormalities in broadly defined domains of psychopathology are bidirectionally associated with metabolic-based medical conditions. This phenotypical convergence is thought to be mediated/moderated by shared pathophysiological mechanisms, including multiple neural systems and peripheral homeostatic systems, as well as to be underlied by common genetic and environmental risk factors.
dysfunction and has been consistently demonstrated to be associated with MDD (Kan et al., 2013). Moreover, two recent clinical trials have reported beneficial effects for adjunctive use of insulin sensitizers (e.g. pioglitazone) in the treatment of MDD, even in the absence of diabetes or metabolic syndrome (Kemp et al., 2012; Sepanjnia et al., 2012). Preliminary evidence indicates that IR prevalence, as assessed by homeostasis model of insulin resistance (HOMA-IR), is also significantly elevated in individuals with BD (Guha et al., 2014). T2DM and IR have been repeatedly shown to impact the brain, as neuroimaging and neurophysiology studies have demonstrated widespread patterns of white matter abnormalities in discrete pathways (van Duinkerken et al., 2012a; Antenor-Dorsey et al., 2013; Reijmer et al., 2013a,b), alterations in functional connectivity (Musen et al., 2012; van Duinkerken et al., 2012b) and in the concentration of brain metabolites (Geissler et al., 2003; Sahin et al., 2008). Similarly to the structural and functional abnormalities in obesity, an interactive effect of diabetes and mood disorders in the alterations in brain metabolites has been reported, as a recent study observed that the levels of NAA and Cr were lowest among individuals with BD and co-morbid T2DM, intermediate in the patients with BD and IR and highest among the euglycemic BD and control subjects (Hajek et al., 2013). More recently, the molecular underpinnings of metabolic dysfunction have been a focus of research. Evidence from longitudinal studies using metabolomic approaches have identified biomarkers that may possibly predate the clinical manifestations, such as obesity, diabetes and CVD, indicating that they may be a more accurate reflection of metabolic status (Wang et al., 2011; Wang-Sattler et al., 2012; Ferrannini et al., 2013). For example, a cohort study found that a combination of the amino-acids isoleucine, leucine, valine, tyrosine, and phenylalanine significantly predicted the
development of diabetes in the follow-up (Wang et al., 2011). Metabolomics methodology has recently been employed in mood disorders and the first studies identified candidate biomarkers that successfully distinguished patients from controls (Sussulini et al., 2009; Zheng et al., 2013). These metabolites include lipids, lipid-metabolism-related molecules (e.g. acetate, choline, and myo-inositol), amino acids (e.g. alanine, valine, leucine, glutamate, glutamine) and energy metabolism-related molecules (e.g. creatine, creatinine) (Sussulini et al., 2009; Zheng et al., 2013). Unsurprisingly, there is an overlap of biomarkers involved in metabolic dysfunction and mood disorders, indicating that integrated metabolomics approaches are a promising tool for further research. Finally, it is important to notice that energy metabolism is tremendously complex, insofar as it is subserved by dynamic and multi-regulated systems, such as insulin-related pathways, adipose-derived and gut hormones, as well as the HPA and the immuno-inflammatory axis. It is recognized that the activation of these systems is not necessarily pathological, as they are an integral part of adaptive physiological mechanisms and allostatic responses. Allostasis refers to broader and more complex modulatory mechanisms, with emphasis on brain control of the primary regulatory processes and on anticipatory physiological and behavioral changes for future events (Schulkin, 2003; Sterling, 2012). Body weight is subjected to allostatic control and weight gain could be perceived as a physiological response, which is certain situations is adaptive (e.g. chronic illnesses) and do not necessarily constitute a dysfunction of the regulatory systems (Tremblay and Chaput, 2012). The existence of the MHO and NWO phenotypes and of the “obesity paradox” indicates that BMI, when used without proper context, may be an inadequate marker, which, in certain specific situations,
98
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
may be even a deceiving one. This observation is of particular interest in chronic diseases of the central nervous system (CNS), such as mood disorders, and should be considered in the interpretation of future studies.
5. Conclusions Emotional and behavioral alterations have been consistently connected to multifaceted metabolic dysfunction. Clinical studies have indicated that not only syndromal mood disorders, as currently defined by diagnostic categories, are associated with energy metabolism abnormalities, but so as more broadly defined neuropsychological constructs, including, but not limited to, response to stress, cognitive function, motivation and reward. Metabolic dysfunction has also been shown to encompass numerous mechanisms that interact with psychopathology, such as glucose and insulin homeostasis, lipids metabolism, sympathoadrenal regulation and the immune-inflammatory axis. Conversely, multiple neural systems that are thought to underlie and possibly bridge both the emotional/behavioral and the metabolic manifestations have been described (Fig. 1). Overall, evidence indicates that the existence of a “metabolic-mood syndrome”, in which the psychopathological and the metabolic manifestations are mechanistically linked and are the predominant clinical presentation, is possible. Empirical and direct evidence are still necessary to support the concepts proposed in this review. To address the issue of heterogeneity in both metabolic and mood disorders, future studies should focus in domains of psychopathology and in multidimensional metabolic dysfunction, as well as in integrated subdomain phenotypes and underlying substrates. The validation of the “metabolic-mood syndrome” concept may have important clinical implications. Several novel interventions have been recently proposed for both obesity and mood disorders and some of those targets metabolism and/or energy-related mechanisms, such as weight-loss interventions, physical exercise, intranasal insulin and insulin sensitizers, anti-inflammatory agents and creatine supplementation (Linde et al., 2011; Chalder et al., 2012; Kemp et al., 2012; Lyoo et al., 2012; McIntyre et al., 2012; Sepanjnia et al., 2012; Busch et al., 2013; Pereira et al., 2013; Raison et al., 2013). Although the clinical trials with these agents have provided promising results, they have been mostly inconsistent and the effect sizes have been modest at best. Therefore, trials with energy-based treatments would greatly benefit from tests in more homogenous populations, insofar as the relationship between mechanism of action/molecular target and pathological processes would be more informed and the likelihood of success would be increased. For example, evidence suggests that BDNF plays a modulator role in the association of metabolic and mood disorders, as a study reported decreased serum BDNF levels in depressed individuals with T2DM when compared to non-depressed diabetics, independently of gender and BMI (Zhou et al., 2013). A separate study testing a weight-loss intervention for obese adults described that reduction in body weight was associated with increase in BDNF (Lee et al., 2014b). In the whole sample, weight reduction was not associated to changes in depression scale scores, but in a subgroup of individuals with greater BDNF increase weight loss was significantly correlated to improvement in depressive symptoms (Lee et al., 2014b), indicating that the positive effect of weight reduction was present only in a specific subgroup of individuals. The emerging field of personalized medicine posits that an individual’s unique physiologic and clinical characteristics are major determinants of vulnerabilities to illnesses and responses to therapies. As discussed in this review, mood and metabolic disorders are extremely heterogeneous and would be better understood through to use of stratified and individualized approaches (Costa e Silva,
2013; Ozomaro et al., 2013). However, in contrast to other areas of medicine in which personalized medicine has been more widely applied, mood and metabolic disorders lack a single defining gene, biomarker, mechanism or even clinical presentation. Within this context, research in this area should consider the complexity and interconnectedness of metabolic systems, both central and peripheral, as well as the impact of compensatory/allostatic responses and individual trajectories. For example, several pharmacological and non-pharmacological weight-loss interventions have reported positive effects of weight reduction in mood symptoms (Simon et al., 2010; Fabricatore et al., 2011; Linde et al., 2011; Busch et al., 2013). Nonetheless, results of clinical trials with the CB1 antagonist rimonabant described a considerable effect in body weight reduction, however, this agent was also significantly associated to increase in levels of anxiety, depression and suicidality (Di Marzo and Despres, 2009; Christopoulou and Kiortsis, 2011). Another study tested a weight-loss program, which consisted of a supervised diet and exercise, reported an increase in depressive symptoms that was correlated to weight reduction and changes in glucose concentrations (Chaput et al., 2008). These results not only add to the accumulating evidence that psychopathology and metabolism are connected, but they also indicate that they should always be analyzed in a contextualized and integrative way. References Abildgaard, A., Solskov, L., Volke, V., Harvey, B.H., Lund, S., Wegener, G., 2011. A highfat diet exacerbates depressive-like behavior in the Flinders Sensitive Line (FSL) rat, a genetic model of depression. Psychoneuroendocrinology 36 (5), 623–633. Adam, E.K., 2006. Transactions among adolescent trait and state emotion and diurnal and momentary cortisol activity in naturalistic settings. Psychoneuroendocrinology 31 (5), 664–679. Afari, N., Noonan, C., Goldberg, J., et al., 2010. Depression and obesity: do shared genes explain the relationship? Depress. Anxiety 27 (9), 799–806. Ahmad, S., Rukh, G., Varga, T.V., et al., 2013. Gene × physical activity interactions in obesity: combined analysis of 111,421 individuals of European ancestry. PLoS Genet. 9 (7), e1003607. Ali, S., Stone, M.A., Peters, J.L., Davies, M.J., Khunti, K., 2006. The prevalence of comorbid depression in adults with type 2 diabetes: a systematic review and metaanalysis. Diabetes Med. 23 (11), 1165–1173. Allender, S., Rayner, M., 2007. The burden of overweight and obesity-related ill health in the UK. Obes. Rev. 8 (5), 467–473. Altman, S., Haeri, S., Cohen, L.J., et al., 2006. Predictors of relapse in bipolar disorder: a review. J. Psychiatr. Pract. 12 (5), 269–282. Anand, A., Barkay, G., Dzemidzic, M., et al., 2011. Striatal dopamine transporter availability in unmedicated bipolar disorder. Bipolar Disord. 13 (4), 406–413. Antenor-Dorsey, J.A., Meyer, E., Rutlin, J., et al., 2013. White matter microstructural integrity in youth with type 1 diabetes. Diabetes 62 (2), 581–589. Ashton, C.H., Moore, P.B., 2011. Endocannabinoid system dysfunction in mood and related disorders. Acta Psychiatr. Scand. 124 (4), 250–261. Atlantis, E., Sullivan, T., 2012. Changes in cardiovascular disease burden associated with psychopathology in Australian adults 2004–2008. Gen. Hosp. Psychiatry 34 (4), 345–351. Baldo, B.A., Kelley, A.E., 2007. Discrete neurochemical coding of distinguishable motivational processes: insights from nucleus accumbens control of feeding. Psychopharmacology (Berl.) 191 (3), 439–459. Barbosa, I.G., Rocha, N.P., de Miranda, A.S., et al., 2012. Increased levels of adipokines in bipolar disorder. J. Psychiatr. Res. 46 (3), 389–393. Barnard, K.D., Skinner, T.C., Peveler, R., 2006. The prevalence of co-morbid depression in adults with type 1 diabetes: systematic literature review. Diabetes Med. 23 (4), 445–448. Baskaran, A., Cha, D.S., Powell, A.M., Jalil, D., McIntyre, R.S., 2014. Sex differences in rates of obesity in bipolar disorder: postulated mechanisms. Bipolar Disord. 16 (1), 83–92. Baxter, A.J., Scott, K.M., Ferrari, A.J., Norman, R.E., Vos, T., Whiteford, H.A., 2014. Challenging the myth of an “epidemic” of common mental disorders: trends in the global prevalence of anxiety and depression between 1990 and 2010. Depress. Anxiety. Bell, C.G., Walley, A.J., Froguel, P., 2005. The genetics of human obesity. Nat. Rev. Genet. 6 (3), 221–234. Bell, M.E., Bhargava, A., Soriano, L., Laugero, K., Akana, S.F., Dallman, M.F., 2002. Sucrose intake and corticosterone interact with cold to modulate ingestive behaviour, energy balance, autonomic outflow and neuroendocrine responses during chronic stress. J. Neuroendocrinol. 14 (4), 330–342. Benard, G., Massa, F., Puente, N., et al., 2012. Mitochondrial CB(1) receptors regulate neuronal energy metabolism. Nat. Neurosci. 15 (4), 558–564. Bennetzen, M.F., Wellner, N., Ahmed, S.S., et al., 2011. Investigations of the human endocannabinoid system in two subcutaneous adipose tissue depots in lean
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104 subjects and in obese subjects before and after weight loss. Int. J. Obes. (Lond.) 35 (11), 1377–1384. Bermudez-Silva, F.J., Cardinal, P., Cota, D., 2012. The role of the endocannabinoid system in the neuroendocrine regulation of energy balance. J. Psychopharmacol. 26 (1), 114–124. Bjornsson, H.T., Fallin, M.D., Feinberg, A.P., 2004. An integrated epigenetic and genetic approach to common human disease. Trends Genet. 20 (8), 350–358. Blaine, B., 2008. Does depression cause obesity? A meta-analysis of longitudinal studies of depression and weight control. J. Health Psychol. 13 (8), 1190–1197. Blond, B.N., Fredericks, C.A., Blumberg, H.P., 2012. Functional neuroanatomy of bipolar disorder: structure, function, and connectivity in an amygdala-anterior paralimbic neural system. Bipolar Disord. 14 (4), 340–355. Bluher, M., Engeli, S., Kloting, N., et al., 2006. Dysregulation of the peripheral and adipose tissue endocannabinoid system in human abdominal obesity. Diabetes 55 (11), 3053–3060. Bocchio-Chiavetto, L., Bagnardi, V., Zanardini, R., et al., 2010. Serum and plasma BDNF levels in major depression: a replication study and meta-analyses. World J. Biol. Psychiatry 11 (6), 763–773. Bond, D.J., Lang, D.J., Noronha, M.M., et al., 2011. The association of elevated body mass index with reduced brain volumes in first-episode mania. Biol. Psychiatry 70 (4), 381–387. Boon, M.R., Kooijman, S., van Dam, A.D., et al., 2014. Peripheral cannabinoid 1 receptor blockade activates brown adipose tissue and diminishes dyslipidemia and obesity. FASEB J. Bourne, C., Aydemir, O., Balanza-Martinez, V., et al., 2013. Neuropsychological testing of cognitive impairment in euthymic bipolar disorder: an individual patient data meta-analysis. Acta Psychiatr. Scand. 128 (3), 149–162. Bowden, C.L., Perlis, R.H., Thase, M.E., et al., 2012. Aims and results of the NIMH systematic treatment enhancement program for bipolar disorder (STEP-BD). CNS Neurosci. Ther. 18 (3), 243–249. Brady Jr., R.O., Cooper, A., Jensen, J.E., et al., 2012. A longitudinal pilot proton MRS investigation of the manic and euthymic states of bipolar disorder. Transl. Psychiatry 2, e160. Brooks, S.J., Nilsson, E.K., Jacobsson, J.A., et al., 2014. BDNF polymorphisms are linked to poorer working memory performance, reduced cerebellar and hippocampal volumes and differences in prefrontal cortex in a Swedish elderly population. PLOS ONE 9 (1), e82707. Brown, E.S., Varghese, F.P., McEwen, B.S., 2004. Association of depression with medical illness: does cortisol play a role? Biol. Psychiatry 55 (1), 1–9. Bruce, D.G., Davis, W.A., Cetrullo, V., Starkstein, S.E., Davis, T.M., 2013. Clinical impact of the temporal relationship between depression and type 2 diabetes: the fremantle diabetes study phase II. PLOS ONE 8 (12), e81254. Buettner, R., Scholmerich, J., Bollheimer, L.C., 2007. High-fat diets: modeling the metabolic disorders of human obesity in rodents. Obesity (Silver Spring) 15 (4), 798–808. Burkhalter, J., Fiumelli, H., Allaman, I., Chatton, J.Y., Martin, J.L., 2003. Brain-derived neurotrophic factor stimulates energy metabolism in developing cortical neurons. J. Neurosci. 23 (23), 8212–8220. Busch, A.M., Whited, M.C., Appelhans, B.M., et al., 2013. Reliable change in depression during behavioral weight loss treatment among women with major depression. Obesity (Silver Spring) 21 (3), E211–E218. Byers, A.L., Vittinghoff, E., Lui, L.Y., et al., 2012. Twenty-year depressive trajectories among older women. Arch. Gen. Psychiatry 69 (10), 1073–1079. Calkin, C., van de Velde, C., Ruzickova, M., et al., 2009. Can body mass index help predict outcome in patients with bipolar disorder? Bipolar Disord. 11 (6), 650–656. Camardese, G., Di Giuda, D., Di Nicola, M., et al., 2014. Imaging studies on dopamine transporter and depression: a review of literature and suggestions for future research. J. Psychiatr. Res. 51, 7–18. Canetti, L., Bachar, E., Berry, E.M., 2002. Food and emotion. Behav. Process. 60 (2), 157–164. Carr, C.P., Martins, C.M., Stingel, A.M., Lemgruber, V.B., Juruena, M.F., 2013. The role of early life stress in adult psychiatric disorders: a systematic review according to childhood trauma subtypes. J. Nerv. Mental Dis. 201 (12), 1007–1020. Cervantes, P., Gelber, S., Kin, F.N., Nair, V.N., Schwartz, G., 2001. Circadian secretion of cortisol in bipolar disorder. J. Psychiatry Neurosci. 26 (5), 411–416. Chalder, M., Wiles, N.J., Campbell, J., et al., 2012. A pragmatic randomised controlled trial to evaluate the cost-effectiveness of a physical activity intervention as a treatment for depression: the treating depression with physical activity (TREAD) trial. Health Technol. Assess. 16 (10), 1–164, iii–iv. Chang, H.H., Yen, S.T., 2012. Association between obesity and depression: evidence from a longitudinal sample of the elderly in Taiwan. Aging Mental Health 16 (2), 173–180. Chang, T.T., Yeh, T.L., Chiu, N.T., et al., 2010. Higher striatal dopamine transporters in euthymic patients with bipolar disorder: a SPECT study with [Tc] TRODAT-1. Bipolar Disord. 12 (1), 102–106. Chaput, J.P., Arguin, H., Gagnon, C., Tremblay, A., 2008. Increase in depression symptoms with weight loss: association with glucose homeostasis and thyroid function. Appl. Physiol. Nutr. Metab. 33 (1), 86–92. Cheng, G., Huang, C., Deng, H., Wang, H., 2012. Diabetes as a risk factor for dementia and mild cognitive impairment: a meta-analysis of longitudinal studies. Intern. Med. J. 42 (5), 484–491. Choi, J., Joseph, L., Pilote, L., 2013. Obesity and C-reactive protein in various populations: a systematic review and meta-analysis. Obes. Rev. 14 (3), 232–244. Chou, K.L., Yu, K.M., 2013. Atypical depressive symptoms and obesity in a national sample of older adults with major depressive disorder. Depress. Anxiety 30 (6), 574–579.
99
Christopoulou, F.D., Kiortsis, D.N., 2011. An overview of the metabolic effects of rimonabant in randomized controlled trials: potential for other cannabinoid 1 receptor blockers in obesity. J. Clin. Pharm. Ther. 36 (1), 10–18. Chu, W.J., Delbello, M.P., Jarvis, K.B., et al., 2013. Magnetic resonance spectroscopy imaging of lactate in patients with bipolar disorder. Psychiatry Res. 213 (3), 230–234. Chuang, J.C., Cui, H., Mason, B.L., et al., 2010. Chronic social defeat stress disrupts regulation of lipid synthesis. J. Lipid Res. 51 (6), 1344–1353. Cizza, G., Ronsaville, D.S., Kleitz, H., et al., 2012. Clinical subtypes of depression are associated with specific metabolic parameters and circadian endocrine profiles in women: the power study. PLoS ONE 7 (1), e28912. Clay, H.B., Sillivan, S., Konradi, C., 2011. Mitochondrial dysfunction and pathology in bipolar disorder and schizophrenia. Int. J. Dev. Neurosci. 29 (3), 311–324. Cole, J.H., Boyle, C.P., Simmons, A., et al., 2013. Body mass index, but not FTO genotype or major depressive disorder, influences brain structure. Neuroscience 252, 109–117. Correll, C.U., Penzner, J.B., Lencz, T., et al., 2007. Early identification and high-risk strategies for bipolar disorder. Bipolar Disord. 9 (4), 324–338. Costa e Silva, J.A., 2013. Personalized medicine in psychiatry: new technologies and approaches. Metabolism 62 (Suppl. 1), S40–S44. Cota, D., Tschop, M.H., Horvath, T.L., Levine, A.S., 2006. Cannabinoids, opioids and eating behavior: the molecular face of hedonism? Brain Res. Rev. 51 (1), 85–107. Cowansage, K.K., LeDoux, J.E., Monfils, M.H., 2010. Brain-derived neurotrophic factor: a dynamic gatekeeper of neural plasticity. Curr. Mol. Pharmacol. 3 (1), 12–29. Czepielewski, L., Daruy-Filho, L., Brietzke, E., Grassi-Oliveira, R., 2013. Bipolar disorder and metabolic syndrome: a systematic review. Rev. Bras. Psiquiatr. 35 (1), 88–93. D’Aquila, P.S., Collu, M., Gessa, G.L., Serra, G., 2000. The role of dopamine in the mechanism of action of antidepressant drugs. Eur. J. Pharmacol. 405 (1–3), 365–373. Daban, C., Vieta, E., Mackin, P., Young, A.H., 2005. Hypothalamic–pituitary–adrenal axis and bipolar disorder. Psychiatr. Clin. N. Am. 28 (2), 469–480. Dave, D.M., Tennant, J., Colman, G., 2011. Isolating the effect of major depression on obesity: role of selection bias. J. Mental Health Policy Econ. 14 (4), 165–186. de Groot, M., Anderson, R., Freedland, K.E., Clouse, R.E., Lustman, P.J., 2001. Association of depression and diabetes complications: a meta-analysis. Psychosomal Med. 63 (4), 619–630. Denis, G.V., Obin, M.S., 2013. ‘Metabolically healthy obesity’: origins and implications. Mol. Asp. Med. 34 (1), 59–70. Despres, J.P., 2001. Health consequences of visceral obesity. Ann. Med. 33 (8), 534–541. Devaux, M., Sassi, F., 2013. Social inequalities in obesity and overweight in 11 OECD countries. Eur. J. Public Health 23 (3), 464–469. Di Marzo, V., Cote, M., Matias, I., et al., 2009. Changes in plasma endocannabinoid levels in viscerally obese men following a 1 year lifestyle modification programme and waist circumference reduction: associations with changes in metabolic risk factors. Diabetologia 52 (2), 213–217. Di Marzo, V., Despres, J.P., 2009. CB1 antagonists for obesity – what lessons have we learned from rimonabant? Nat. Rev. Endocrinol. 5 (11), 633–638. Diener, C., Kuehner, C., Brusniak, W., Ubl, B., Wessa, M., Flor, H., 2012. A metaanalysis of neurofunctional imaging studies of emotion and cognition in major depression. Neuroimage 61 (3), 677–685. Dillon, D.G., Rosso, I.M., Pechtel, P., Killgore, W.D., Rauch, S.L., Pizzagalli, D.A., 2014. Peril and pleasure: an RDoC-inspired examination of threat responses and reward processing in anxiety and depression. Depress. Anxiety 31 (3), 233–249. Dincheva, I., Glatt, C.E., Lee, F.S., 2012. Impact of the BDNF Val66Met polymorphism on cognition: implications for behavioral genetics. Neuroscientist 18 (5), 439–451. Dlugos, A., Childs, E., Stuhr, K.L., Hillard, C.J., de Wit, H., 2012. Acute stress increases circulating anandamide and other N-acylethanolamines in healthy humans. Neuropsychopharmacology 37 (11), 2416–2427. Dong, Q., Liu, J.J., Zheng, R.Z., et al., 2013. Obesity and depressive symptoms in the elderly: a survey in the rural area of Chizhou, Anhui province. Int. J. Geriatr. Psychiatry 28 (3), 227–232. Dowlati, Y., Herrmann, N., Swardfager, W., et al., 2010. A meta-analysis of cytokines in major depression. Biol. Psychiatry 67 (5), 446–457. Elder, C.R., Gullion, C.M., Funk, K.L., Debar, L.L., Lindberg, N.M., Stevens, V.J., 2012. Impact of sleep, screen time, depression and stress on weight change in the intensive weight loss phase of the LIFE study. Int. J. Obes. (Lond.) 36 (1), 86–92. Everson, S.A., Maty, S.C., Lynch, J.W., Kaplan, G.A., 2002. Epidemiologic evidence for the relation between socioeconomic status and depression, obesity, and diabetes. J. Psychosomal Res. 53 (4), 891–895. Fabricatore, A.N., Wadden, T.A., Higginbotham, A.J., et al., 2011. Intentional weight loss and changes in symptoms of depression: a systematic review and metaanalysis. Int. J. Obes. (Lond.) 35 (11), 1363–1376. Fagiolini, A., Frank, E., Scott, J.A., Turkin, S., Kupfer, D.J., 2005. Metabolic syndrome in bipolar disorder: findings from the Bipolar Disorder Center for Pennsylvanians. Bipolar Disord. 7 (5), 424–430. Fernandes, B.S., Gama, C.S., Cereser, K.M., et al., 2011. Brain-derived neurotrophic factor as a state-marker of mood episodes in bipolar disorders: a systematic review and meta-regression analysis. J. Psychiatr. Res. 45 (8), 995–1004. Ferrannini, E., Natali, A., Bell, P., Cavallo-Perin, P., Lalic, N., Mingrone, G., 1997. Insulin resistance and hypersecretion in obesity. European Group for the Study of Insulin Resistance (EGIR). J. Clin. Invest. 100 (5), 1166–1173. Ferrannini, E., Natali, A., Camastra, S., et al., 2013. Early metabolic markers of the development of dysglycemia and type 2 diabetes and their physiological significance. Diabetes 62 (5), 1730–1737.
100
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Fesinmeyer, M.D., North, K.E., Ritchie, M.D., et al., 2013. Genetic risk factors for BMI and obesity in an ethnically diverse population: results from the population architecture using genomics and epidemiology (PAGE) study. Obesity (Silver Spring) 21 (4), 835–846. Field, A.E., Camargo Jr., C.A., Ogino, S., 2013. The merits of subtyping obesity: one size does not fit all. JAMA 310 (20), 2147–2148. Finkelstein, E.A., Brown, D.S., Wrage, L.A., Allaire, B.T., Hoerger, T.J., 2010. Individual and aggregate years-of-life-lost associated with overweight and obesity. Obesity (Silver Spring) 18 (2), 333–339. Finkelstein, E.A., Trogdon, J.G., Cohen, J.W., Dietz, W., 2009. Annual medical spending attributable to obesity: payer-and service-specific estimates. Health Aff. (Millwood) 28 (5), w822–w831. Flegal, K.M., Kit, B.K., Orpana, H., Graubard, B.I., 2013. Association of all-cause mortality with overweight and obesity using standard body mass index categories: a systematic review and meta-analysis. JAMA 309 (1), 71–82. Flint, J., Kendler, K.S., 2014. The genetics of major depression. Neuron 81 (3), 484–503. Forde, N.J., Ronan, L., Suckling, J., et al., 2014. Structural neuroimaging correlates of allelic variation of the BDNF val66met polymorphism. Neuroimage 90, 280–289. Fuemmeler, B.F., Agurs-Collins, T., McClernon, F.J., Kollins, S.H., Garrett, M.E., AshleyKoch, A.E., 2009. Interactions between genotype and depressive symptoms on obesity. Behav. Genet. 39 (3), 296–305. Galler, J.R., Bryce, C.P., Waber, D., et al., 2010. Early childhood malnutrition predicts depressive symptoms at ages 11–17. J. Child Psychol. Psychiatry 51 (7), 789–798. Garcia-Garcia, I., Jurado, M.A., Garolera, M., et al., 2012. Functional connectivity in obesity during reward processing. Neuroimage 66C, 232–239. Gardner, A., Boles, R.G., 2011. Beyond the serotonin hypothesis: mitochondria, inflammation and neurodegeneration in major depression and affective spectrum disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 35 (3), 730–743. Gault, V.A., Porter, W.D., Flatt, P.R., Holscher, C., 2010. Actions of exendin-4 therapy on cognitive function and hippocampal synaptic plasticity in mice fed a high-fat diet. Int. J. Obes. (Lond.) 34 (8), 1341–1344. Gazdzinski, S., Kornak, J., Weiner, M.W., Meyerhoff, D.J., 2008. Body mass index and magnetic resonance markers of brain integrity in adults. Ann. Neurol. 63 (5), 652–657. Gazdzinski, S., Millin, R., Kaiser, L.G., et al., 2010. BMI and neuronal integrity in healthy, cognitively normal elderly: a proton magnetic resonance spectroscopy study. Obesity (Silver Spring) 18 (4), 743–748. Geissler, A., Frund, R., Scholmerich, J., Feuerbach, S., Zietz, B., 2003. Alterations of cerebral metabolism in patients with diabetes mellitus studied by proton magnetic resonance spectroscopy. Exp. Clin. Endocrinol. Diabetes 111 (7), 421–427. Glaus, J., Vandeleur, C., Gholam-Rezaee, M., et al., 2012. Atypical depression and alcohol misuse are related to the cardiovascular risk in the general population. Acta Psychiatr. Scand. Gloy, V.L., Briel, M., Bhatt, D.L., et al., 2013. Bariatric surgery versus non-surgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. BMJ 347, f5934. Gluckman, P.D., Hanson, M.A., Cooper, C., Thornburg, K.L., 2008. Effect of in utero and early-life conditions on adult health and disease. N. Engl. J. Med. 359 (1), 61–73. Gobbi, G., Bambico, F.R., Mangieri, R., et al., 2005. Antidepressant-like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc. Natl. Acad. Sci. U. S. A. 102 (51), 18620–18625. Gold, P.W., Chrousos, G.P., 2002. Organization of the stress system and its dysregulation in melancholic and atypical depression: high vs low CRH/NE states. Mol. Psychiatry 7 (3), 254–275. Gold, S.M., Dziobek, I., Sweat, V., et al., 2007. Hippocampal damage and memory impairments as possible early brain complications of type 2 diabetes. Diabetologia 50 (4), 711–719. Golden, S.H., Lazo, M., Carnethon, M., et al., 2008. Examining a bidirectional association between depressive symptoms and diabetes. JAMA 299 (23), 2751–2759. Goldstein, B.I., Liu, S.M., Schaffer, A., Sala, R., Blanco, C., 2013. Obesity and the threeyear longitudinal course of bipolar disorder. Bipolar Disord. 15 (3), 284–293. Goldstein, B.I., Liu, S.M., Zivkovic, N., Schaffer, A., Chien, L.C., Blanco, C., 2011. The burden of obesity among adults with bipolar disorder in the United States. Bipolar Disord. 13 (4), 387–395. Gonzalez de Castro, D., Clarke, P.A., Al-Lazikani, B., Workman, P., 2013. Personalized cancer medicine: molecular diagnostics, predictive biomarkers, and drug resistance. Clin. Pharmacol. Ther. 93 (3), 252–259. Goodman, E., Whitaker, R.C., 2002. A prospective study of the role of depression in the development and persistence of adolescent obesity. Pediatrics 110 (3), 497–504. Grigoriadis, S., VonderPorten, E.H., Mamisashvili, L., et al., 2013. The impact of maternal depression during pregnancy on perinatal outcomes: a systematic review and meta-analysis. J Clin Psychiatry 74 (4), e321–e341. Guenard, F., Deshaies, Y., Cianflone, K., Kral, J.G., Marceau, P., Vohl, M.C., 2013a. Differential methylation in glucoregulatory genes of offspring born before vs. after maternal gastrointestinal bypass surgery. Proc. Natl. Acad. Sci. U. S. A. 110 (28), 11439–11444. Guenard, F., Tchernof, A., Deshaies, Y., et al., 2013b. Methylation and expression of immune and inflammatory genes in the offspring of bariatric bypass surgery patients. J. Obes., 492170. Guha, P., Bhowmick, K., Mazumder, P., Ghosal, M., Chakraborty, I., Burman, P., 2014. Assessment of insulin resistance and metabolic syndrome in drug naive patients of bipolar disorder. Indian J. Clin. Biochem. 29 (1), 51–56.
Gunstad, J., Lhotsky, A., Wendell, C.R., Ferrucci, L., Zonderman, A.B., 2010. Longitudinal examination of obesity and cognitive function: results from the Baltimore longitudinal study of aging. Neuroepidemiology 34 (4), 222–229. Gunstad, J., Paul, R.H., Cohen, R.A., Tate, D.F., Spitznagel, M.B., Gordon, E., 2007. Elevated body mass index is associated with executive dysfunction in otherwise healthy adults. Compr. Psychiatry 48 (1), 57–61. Gunstad, J., Paul, R.H., Spitznagel, M.B., et al., 2006. Exposure to early life trauma is associated with adult obesity. Psychiatry Res. 142 (1), 31–37. Haack, M., Hinze-Selch, D., Fenzel, T., et al., 1999. Plasma levels of cytokines and soluble cytokine receptors in psychiatric patients upon hospital admission: effects of confounding factors and diagnosis. J. Psychiatr. Res. 33 (5), 407–418. Hajek, T., Calkin, C., Blagdon, R., Slaney, C., Alda, M., 2013. Type 2 diabetes mellitus: a potentially modifiable risk factor for neurochemical brain changes in bipolar disorders. Biol. Psychiatry. Hamer, M., Batty, G.D., Kivimaki, M., 2011. Haemoglobin A1c, fasting glucose and future risk of elevated depressive symptoms over 2 years of follow-up in the English Longitudinal Study of Ageing. Psychol. Med. 41 (9), 1889–1896. Hamer, M., Batty, G.D., Kivimaki, M., 2012. Risk of future depression in people who are obese but metabolically healthy: the English longitudinal study of ageing. Mol. Psychiatry 17 (9), 940–945. Harald, B., Gordon, P., 2012. Meta-review of depressive subtyping models. J. Affect. Disord. 139 (2), 126–140. Hebebrand, J., Friedel, S., Schauble, N., Geller, F., Hinney, A., 2003. Perspectives: molecular genetic research in human obesity. Obes. Rev. 4 (3), 139–146. Hickman, R.J., Khambaty, T., Stewart, J.C., 2013. C-reactive protein is elevated in atypical but not nonatypical depression: data from the National Health and Nutrition Examination Survey (NHANES) 1999–2004. J. Behav. Med. Hill, M.N., Gorzalka, B.B., 2009. The endocannabinoid system and the treatment of mood and anxiety disorders. CNS Neurol. Disord. Drug Targets 8 (6), 451–458. Hill, M.N., Ho, W.S., Hillard, C.J., Gorzalka, B.B., 2008. Differential effects of the antidepressants tranylcypromine and fluoxetine on limbic cannabinoid receptor binding and endocannabinoid contents. J. Neural. Transm. 115 (12), 1673–1679. Hill, M.N., McEwen, B.S., 2010. Involvement of the endocannabinoid system in the neurobehavioural effects of stress and glucocorticoids. Prog. Neuropsychopharmacol. Biol. Psychiatry 34 (5), 791–797. Hill, M.N., Patel, S., Campolongo, P., Tasker, J.G., Wotjak, C.T., Bains, J.S., 2010. Functional interactions between stress and the endocannabinoid system: from synaptic signaling to behavioral output. J. Neurosci. 30 (45), 14980–14986. Hill, M.N., Tasker, J.G., 2012. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic–pituitary–adrenal axis. Neuroscience 204, 5–16. Hitze, B., Hubold, C., van Dyken, R., et al., 2010. How the selfish brain organizes its supply and demand. Front. Neuroenergetics 2, 7. Ho, B.C., Milev, P., O’Leary, D.S., Librant, A., Andreasen, N.C., Wassink, T.H., 2006. Cognitive and magnetic resonance imaging brain morphometric correlates of brain-derived neurotrophic factor Val66Met gene polymorphism in patients with schizophrenia and healthy volunteers. Arch. Gen. Psychiatry 63 (7), 731–740. Hong, C.J., Liou, Y.J., Tsai, S.J., 2011. Effects of BDNF polymorphisms on brain function and behavior in health and disease. Brain Res. Bull. 86 (5–6), 287–297. Horesh, N., Apter, A., Zalsman, G., 2011. Timing, quantity and quality of stressful life events in childhood and preceding the first episode of bipolar disorder. J. Affect. Disord. 134 (1–3), 434–437. Horesh, N., Iancu, I., 2010. A comparison of life events in patients with unipolar disorder or bipolar disorder and controls. Compr. Psychiatry 51 (2), 157–164. Howren, M.B., Lamkin, D.M., Suls, J., 2009. Associations of depression with C-reactive protein IL-1, and IL-6: a meta-analysis. Psychosomal Med. 71 (2), 171–186. Hu, H., Xu, Y., Liu, C., Zhao, H., Zhang, H., Wang, L., 2014. Changes in behavior and in brain glucose metabolism in rats after nine weeks on a high fat diet: a randomized controlled trial. Shanghai Arch. Psychiatry 26 (3), 129–137. Hussain, S.S., Bloom, S.R., 2013. The regulation of food intake by the gut–brain axis: implications for obesity. Int. J. Obes. (Lond.) 37 (5), 625–633. Iacobellis, G., Ribaudo, M.C., Zappaterreno, A., Iannucci, C.V., Leonetti, F., 2005. Prevalence of uncomplicated obesity in an Italian obese population. Obes. Res. 13 (6), 1116–1122. Iio, W., Tokutake, Y., Koike, H., et al., 2014. Effects of chronic mild food restriction on behavior and the hypothalamic malonyl-CoA signaling pathway. Anim. Sci. J. Iio, W., Tokutake, Y., Matsukawa, N., Tsukahara, T., Chohnan, S., Toyoda, A., 2012. Anorexic behavior and elevation of hypothalamic malonyl-CoA in socially defeated rats. Biochem. Biophys. Res. Commun. 421 (2), 301–304. Insel, T.R., 2014. The NIMH Research Domain Criteria (RDoC) project: precision medicine for psychiatry. Am. J. Psychiatry 171 (4), 395–397. Iosifescu, D.V., Renshaw, P.E., 2003. 31P-magnetic resonance spectroscopy and thyroid hormones in major depressive disorder: toward a bioenergetic mechanism in depression? Harv. Rev. Psychiatry 11 (2), 51–63. Jackson, M.B., Ahima, R.S., 2006. Neuroendocrine and metabolic effects of adipocytederived hormones. Clin. Sci. (Lond.) 110 (2), 143–152. Jauch-Chara, K., Oltmanns, K.M., 2014. Obesity – a neuropsychological disease? Systematic review and neuropsychological model. Prog. Neurobiol. 114, 84–101. Jialin, W., Yi, Z., Weijie, Y., 2012. Relationship between body mass index and mortality in hemodialysis patients: a meta-analysis. Nephron Clin. Pract. 121 (3–4), c102–c111. Jokela, M., Elovainio, M., Keltikangas-Jarvinen, L., et al., 2012. Body mass index and depressive symptoms: instrumental-variables regression with genetic risk score. Genes Brain Behav.
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104 Jokela, M., Hamer, M., Singh-Manoux, A., Batty, G.D., Kivimaki, M., 2013. Association of metabolically healthy obesity with depressive symptoms: pooled analysis of eight studies. Mol. Psychiatry. Jones, A., McMillan, M.R., Jones, R.W., et al., 2012. Adiposity is associated with blunted cardiovascular, neuroendocrine and cognitive responses to acute mental stress. PLoS ONE 7 (6), e39143. Kahn, S.E., Hull, R.L., Utzschneider, K.M., 2006. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444 (7121), 840–846. Kan, C., Silva, N., Golden, S.H., et al., 2013. A systematic review and meta-analysis of the association between depression and insulin resistance. Diabetes Care 36 (2), 480–489. Karatsoreos, I.N., Thaler, J.P., Borgland, S.L., Champagne, F.A., Hurd, Y.L., Hill, M.N., 2013. Food for thought: hormonal, experiential, and neural influences on feeding and obesity. J. Neurosci. 33 (45), 17610–17616. Karelis, A.D., Faraj, M., Bastard, J.P., et al., 2005. The metabolically healthy but obese individual presents a favorable inflammation profile. J. Clin. Endocrinol. Metab. 90 (7), 4145–4150. Kelly, T., Yang, W., Chen, C.S., Reynolds, K., He, J., 2008. Global burden of obesity in 2005 and projections to 2030. Int. J. Obes. (Lond.) 32 (9), 1431–1437. Kemp, D.E., Gao, K., Chan, P.K., Ganocy, S.J., Findling, R.L., Calabrese, J.R., 2010. Medical comorbidity in bipolar disorder: relationship between illnesses of the endocrine/metabolic system and treatment outcome. Bipolar Disord. 12 (4), 404–413. Kemp, D.E., Ismail-Beigi, F., Ganocy, S.J., et al., 2012. Use of insulin sensitizers for the treatment of major depressive disorder: a pilot study of pioglitazone for major depression accompanied by abdominal obesity. J. Affect. Disord. 136 (3), 1164–1173. Kemp, D.E., Sylvia, L.G., Calabrese, J.R., et al., 2014. General medical burden in bipolar disorder: findings from the LiTMUS comparative effectiveness trial. Acta Psychiatr. Scand. 129 (1), 24–34. Kendler, K.S., Aggen, S.H., Neale, M.C., 2013. Evidence for multiple genetic factors underlying DSM-IV criteria for major depression. JAMA Psychiatry 70 (6), 599–607. Kendler, K.S., Gatz, M., Gardner, C.O., Pedersen, N.L., 2006. A Swedish national twin study of lifetime major depression. Am. J. Psychiatry 163 (1), 109–114. Keranen, A.M., Rasinaho, E., Hakko, H., Savolainen, M., Lindeman, S., 2010. Eating behavior in obese and overweight persons with and without anhedonia. Appetite 55 (3), 726–729. Kerner, B., 2014. Genetics of bipolar disorder. Appl. Clin. Genet. 7, 33–42. Kessler, R.C., Berglund, P., Demler, O., Jin, R., Merikangas, K.R., Walters, E.E., 2005. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62 (6), 593–602. Khan, A., Schwartz, K.A., Kolts, R.L., Brown, W.A., 2007. BMI, sex, and antidepressant response. J. Affect. Disord. 99 (1–3), 101–106. Kim, E.Y., Miklowitz, D.J., Biuckians, A., Mullen, K., 2007. Life stress and the course of early-onset bipolar disorder. J. Affect. Disord. 99 (1–3), 37–44. Kim, M., Bae, S., Lim, K.M., 2013. Impact of high fat diet-induced obesity on the plasma levels of monoamine neurotransmitters in C57BL/6 mice. Biomol. Ther. (Seoul) 21 (6), 476–480. Kloiber, S., Ising, M., Reppermund, S., et al., 2007. Overweight and obesity affect treatment response in major depression. Biol. Psychiatry 62 (4), 321–326. Koethe, D., Llenos, I.C., Dulay, J.R., et al., 2007. Expression of CB1 cannabinoid receptor in the anterior cingulate cortex in schizophrenia, bipolar disorder, and major depression. J. Neural Transm. 114 (8), 1055–1063. Kraguljac, N.V., Reid, M., White, D., et al., 2012. Neurometabolites in schizophrenia and bipolar disorder – a systematic review and meta-analysis. Psychiatry Res. 203 (2–3), 111–125. Kramer, C.K., Zinman, B., Retnakaran, R., 2013. Are metabolically healthy overweight and obesity benign conditions? A systematic review and meta-analysis. Ann. Intern. Med. 159 (11), 758–769. Kubera, B., Hubold, C., Zug, S., et al., 2012. The brain’s supply and demand in obesity. Front. Neuroenergetics 4, 4. Kullmann, S., Heni, M., Veit, R., et al., 2012. The obese brain: association of body mass index and insulin sensitivity with resting state network functional connectivity. Hum. Brain Mapp. 33 (5), 1052–1061. Kuswanto, C.N., Sum, M.Y., Yang, G.L., Nowinski, W.L., McIntyre, R.S., Sim, K., 2014. Increased body mass index makes an impact on brain white-matter integrity in adults with remitted first-episode mania. Psychol. Med. 44 (3), 533–541. Kyrou, I., Chrousos, G.P., Tsigos, C., 2006. Stress, visceral obesity, and metabolic complications. Ann. N. Y. Acad. Sci. 1083, 77–110. Lai, J.S., Hiles, S., Bisquera, A., Hure, A.J., McEvoy, M., Attia, J., 2014. A systematic review and meta-analysis of dietary patterns and depression in communitydwelling adults. Am. J. Clin. Nutr. 99 (1), 181–197. Lamers, F., de Jonge, P., Nolen, W.A., et al., 2010. Identifying depressive subtypes in a large cohort study: results from the Netherlands Study of Depression and Anxiety (NESDA). J. Clin. Psychiatry 71 (12), 1582–1589. Lamers, F., Vogelzangs, N., Merikangas, K.R., de Jonge, P., Beekman, A.T., Penninx, B.W., 2013. Evidence for a differential role of HPA-axis function, inflammation and metabolic syndrome in melancholic versus atypical depression. Mol. Psychiatry 18 (6), 692–699. Le Foll, B., Gorelick, D.A., Goldberg, S.R., 2009. The future of endocannabinoidoriented clinical research after CB1 antagonists. Psychopharmacology (Berl.) 205 (1), 171–174. Le, T.K., Curtis, B., Kahle-Wrobleski, K., Johnston, J., Haldane, D., Melfi, C., 2011. Treatment patterns and resource use among patients with comorbid diabetes mellitus and major depressive disorder. J. Med. Econ. 14 (4), 440–447.
101
Lee, C., Tsenkova, V., Carr, D., 2014a. Childhood trauma and metabolic syndrome in men and women. Soc. Sci. Med. 105, 122–130. Lee, I.T., Fu, C.P., Lee, W.J., et al., 2014b. Brain-derived neurotrophic factor, but not body weight, correlated with a reduction in depression scale scores in men with metabolic syndrome: a prospective weight-reduction study. Diabetol. Metab. Syndr. 6 (1), 18. Lee, R.S., Hermens, D.F., Porter, M.A., Redoblado-Hodge, M.A., 2012. A meta-analysis of cognitive deficits in first-episode Major Depressive Disorder. J. Affect. Disord. 140 (2), 113–124. Legenbauer, T., De Zwaan, M., Benecke, A., Muhlhans, B., Petrak, F., Herpertz, S., 2009. Depression and anxiety: their predictive function for weight loss in obese individuals. Obes. Facts 2 (4), 227–234. Legenbauer, T., Petrak, F., de Zwaan, M., Herpertz, S., 2011. Influence of depressive and eating disorders on short- and long-term course of weight after surgical and nonsurgical weight loss treatment. Compr. Psychiatry 52 (3), 301–311. Levitan, R.D., Davis, C., Kaplan, A.S., Arenovich, T., Phillips, D.I., Ravindran, A.V., 2012. Obesity comorbidity in unipolar major depressive disorder: refining the core phenotype. J. Clin. Psychiatry 73 (8), 1119–1124. Liao, X., Lochhead, P., Nishihara, R., et al., 2012. Aspirin use, tumor PIK3CA mutation, colorectal-cancer survival. N. Engl. J. Med. 367 (17), 1596–1606. Liao, Y., Huang, X., Wu, Q., et al., 2013. Is depression a disconnection syndrome? Meta-analysis of diffusion tensor imaging studies in patients with MDD. J. Psychiatry Neurosci. 38 (1), 49–56. Lin, E.J., Sun, M., Choi, E.Y., Magee, D., Stets, C.W., During, M.J., 2014. Social overcrowding as a chronic stress model that increases adiposity in mice. Psychoneuroendocrinology 51C, 318–330. Lin, F., Weng, S., Xie, B., Wu, G., Lei, H., 2011. Abnormal frontal cortex white matter connections in bipolar disorder: a DTI tractography study. J. Affect. Disord. 131 (1–3), 299–306. Linde, J.A., Simon, G.E., Ludman, E.J., et al., 2011. A randomized controlled trial of behavioral weight loss treatment versus combined weight loss/depression treatment among women with comorbid obesity and depression. Ann. Behav. Med. 41 (1), 119–130. Lopresti, A.L., Hood, S.D., Drummond, P.D., 2013. A review of lifestyle factors that contribute to important pathways associated with major depression: diet, sleep and exercise. J. Affect. Disord. 148 (1), 12–27. Lu, X.Y., 2007. The leptin hypothesis of depression: a potential link between mood disorders and obesity? Curr. Opin. Pharmacol. 7 (6), 648–652. Lu, Y., Hajifathalian, K., Ezzati, M., Woodward, M., Rimm, E.B., Danaei, G., 2014. Metabolic mediators of the effects of body-mass index, overweight, and obesity on coronary heart disease and stroke: a pooled analysis of 97 prospective cohorts with 1.8 million participants. Lancet 383 (9921), 970–983. Ludescher, B., Machann, J., Eschweiler, G.W., et al., 2011. Active depression is associated with regional adiposity in the upper abdomen and the neck. Int. J. Psychiatry Med. 41 (3), 271–280. Luppino, F.S., de Wit, L.M., Bouvy, P.F., et al., 2010. Overweight, obesity, and depression: a systematic review and meta-analysis of longitudinal studies. Arch. Gen. Psychiatry 67 (3), 220–229. Lustman, P.J., Anderson, R.J., Freedland, K.E., de Groot, M., Carney, R.M., Clouse, R.E., 2000. Depression and poor glycemic control: a meta-analytic review of the literature. Diabetes Care 23 (7), 934–942. Lyoo, I.K., Yoon, S., Kim, T.S., et al., 2012. A randomized, double-blind placebocontrolled trial of oral creatine monohydrate augmentation for enhanced response to a selective serotonin reuptake inhibitor in women with major depressive disorder. Am. J. Psychiatry 169 (9), 937–945. Manji, H., Kato, T., Di Prospero, N.A., et al., 2012. Impaired mitochondrial function in psychiatric disorders. Nat. Rev. Neurosci. 13 (5), 293–307. Mann-Wrobel, M.C., Carreno, J.T., Dickinson, D., 2011. Meta-analysis of neuropsychological functioning in euthymic bipolar disorder: an update and investigation of moderator variables. Bipolar Disord. 13 (4), 334–342. Marazziti, D., Baroni, S., Picchetti, M., et al., 2011. Mitochondrial alterations and neuropsychiatric disorders. Curr. Med. Chem. 18 (30), 4715–4721. Marijnissen, R.M., Bus, B.A., Holewijn, S., et al., 2011. Depressive symptom clusters are differentially associated with general and visceral obesity. J. Am. Geriatr. Soc. 59 (1), 67–72. Markham, A., Cameron, I., Bains, R., et al., 2012. Brain-derived neurotrophic factor-mediated effects on mitochondrial respiratory coupling and neuroprotection share the same molecular signalling pathways. Eur. J. Neurosci. 35 (3), 366–374. Marosi, K., Mattson, M.P., 2014. BDNF mediates adaptive brain and body responses to energetic challenges. Trends Endocrinol. Metab. 25 (2), 89–98. Marques-Vidal, P., Pecoud, A., Hayoz, D., et al., 2008. Prevalence of normal weight obesity in Switzerland: effect of various definitions. Eur. J. Nutr. 47 (5), 251–257. Mather, A.A., Cox, B.J., Enns, M.W., Sareen, J., 2009. Associations of obesity with psychiatric disorders and suicidal behaviors in a nationally representative sample. J. Psychosomal Res. 66 (4), 277–285. Mathers, C.D., Lopez, A.D., Murray, C.J.L., 2006. The Burden of Disease and Mortality by Condition: Data, Methods, and Results for 2001. Matias, I., Gatta-Cherifi, B., Tabarin, A., et al., 2012. Endocannabinoids measurement in human saliva as potential biomarker of obesity. PLoS ONE 7 (7), e42399. McCrimmon, R.J., Ryan, C.M., Frier, B.M., 2012. Diabetes and cognitive dysfunction. Lancet 379 (9833), 2291–2299. McIntyre, R.S., Danilewitz, M., Liauw, S.S., et al., 2010. Bipolar disorder and metabolic syndrome: an international perspective. J. Affect. Disord. 126 (3), 366–387. McIntyre, R.S., Muzina, D.J., Kemp, D.E., et al., 2008. Bipolar disorder and suicide: research synthesis and clinical translation. Curr. Psychiatry Rep. 10 (1), 66–72.
102
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
McIntyre, R.S., Soczynska, J.K., Konarski, J.Z., et al., 2007. Should depressive syndromes be reclassified as “metabolic syndrome type II”? Ann. Clin. Psychiatry 19 (4), 257–264. McIntyre, R.S., Soczynska, J.K., Woldeyohannes, H.O., et al., 2012. A randomized, double-blind, controlled trial evaluating the effect of intranasal insulin on neurocognitive function in euthymic patients with bipolar disorder. Bipolar Disord. 14 (7), 697–706. Midei, A.J., Matthews, K.A., Bromberger, J.T., 2010. Childhood abuse is associated with adiposity in midlife women: possible pathways through trait anger and reproductive hormones. Psychosomal Med. 72 (2), 215–223. Midei, A.J., Matthews, K.A., Chang, Y.F., Bromberger, J.T., 2013. Childhood physical abuse is associated with incident metabolic syndrome in mid-life women. Health Psychol. 32 (2), 121–127. Milaneschi, Y., Simonsick, E.M., Vogelzangs, N., et al., 2012. Leptin, abdominal obesity, and onset of depression in older men and women. J. Clin. Psychiatry 73 (9), 1205–1211. Milaneschi, Y., Sutin, A.R., Terracciano, A., et al., 2014. The association between leptin and depressive symptoms is modulated by abdominal adiposity. Psychoneuroendocrinology 42, 1–10. Mitchell, P.B., Morris, M.J., 2007. Depression and anxiety with rimonabant. Lancet 370 (9600), 1671–1672. Moore, T.R., 2010. Fetal exposure to gestational diabetes contributes to subsequent adult metabolic syndrome. Am. J. Obstet. Gynecol. 202 (6), 643–649. Moreira, F.A., Kaiser, N., Monory, K., Lutz, B., 2008. Reduced anxiety-like behaviour induced by genetic and pharmacological inhibition of the endocannabinoiddegrading enzyme fatty acid amide hydrolase (FAAH) is mediated by CB1 receptors. Neuropharmacology 54 (1), 141–150. Morkedal, B., Vatten, L.J., Romundstad, P.R., Laugsand, L.E., Janszky, I., 2014. Risk of myocardial infarction and heart failure among metabolically healthy but obese individuals: HUNT (Nord-Trondelag Health Study), Norway. J. Am. Coll. Cardiol. 63 (11), 1071–1078. Morris, S.E., Cuthbert, B.N., 2012. Research domain criteria: cognitive systems, neural circuits, and dimensions of behavior. Dialogues Clin. Neurosci. 14 (1), 29–37. Moylan, S., Maes, M., Wray, N.R., Berk, M., 2013. The neuroprogressive nature of major depressive disorder: pathways to disease evolution and resistance, and therapeutic implications. Mol. Psychiatry 18 (5), 595–606. Mujica-Parodi, L.R., Renelique, R., Taylor, M.K., 2009. Higher body fat percentage is associated with increased cortisol reactivity and impaired cognitive resilience in response to acute emotional stress. Int. J. Obes. (Lond.) 33 (1), 157–165. Munkholm, K., Vinberg, M., Vedel Kessing, L., 2013. Cytokines in bipolar disorder: a systematic review and meta-analysis. J. Affect. Disord. 144 (1–2), 16–27. Musen, G., Jacobson, A.M., Bolo, N.R., et al., 2012. Resting-state brain functional connectivity is altered in type 2 diabetes. Diabetes 61 (9), 2375–2379. Nanni, V., Uher, R., Danese, A., 2012. Childhood maltreatment predicts unfavorable course of illness and treatment outcome in depression: a meta-analysis. Am. J. Psychiatry 169 (2), 141–151. Naukkarinen, J., Rissanen, A., Kaprio, J., Pietilainen, K.H., 2012. Causes and consequences of obesity: the contribution of recent twin studies. Int. J. Obes. (Lond.) 36 (8), 1017–1024. Nierenberg, A.A., Husain, M.M., Trivedi, M.H., et al., 2010. Residual symptoms after remission of major depressive disorder with citalopram and risk of relapse: a STAR*D report. Psychol. Med. 40 (1), 41–50. Nomura, Y., Wickramaratne, P.J., Pilowsky, D.J., et al., 2007. Low birth weight and risk of affective disorders and selected medical illness in offspring at high and low risk for depression. Compr. Psychiatry 48 (5), 470–478. OECD, 2013. Health at a Glance 2013: OECD Indicators. OECD Publishing, http://dx.doi.org/10.1787/health glance-2013-en. Ohsiek, S., Williams, M., 2011. Psychological factors influencing weight loss maintenance: an integrative literature review. J. Am. Acad. Nurse Pract. 23 (11), 592–601. Oliver, G., Wardle, J., Gibson, E.L., 2000. Stress and food choice: a laboratory study. Psychosomal Med. 62 (6), 853–865. Oliveros, E., Somers, V.K., Sochor, O., Goel, K., Lopez-Jimenez, F., 2014. The concept of normal weight obesity. Prog. Cardiovasc. Dis. 56 (4), 426–433. Oreopoulos, A., Padwal, R., Kalantar-Zadeh, K., Fonarow, G.C., Norris, C.M., McAlister, F.A., 2008. Body mass index and mortality in heart failure: a meta-analysis. Am. Heart J. 156 (1), 13–22. Oskooilar, N., Wilcox, C.S., Tong, M.L., Grosz, D.E., 2009. Body mass index and response to antidepressants in depressed research subjects. J. Clin. Psychiatry 70 (11), 1609–1610. Overstreet, D.H., Friedman, E., Mathe, A.A., Yadid, G., 2005. The Flinders Sensitive Line rat: a selectively bred putative animal model of depression. Neurosci. Biobehav. Rev. 29 (4–5), 739–759. Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch. Gen. Psychiatry 63 (5), 530–538. Ozomaro, U., Wahlestedt, C., Nemeroff, C.B., 2013. Personalized medicine in psychiatry: problems and promises. BMC Med. 11, 132. Padwal, R., McAlister, F.A., McMurray, J.J., et al., 2013. The obesity paradox in heart failure patients with preserved versus reduced ejection fraction: a meta-analysis of individual patient data. Int. J. Obes. (Lond.). Pagoto, S., Schneider, K.L., Whited, M.C., et al., 2013. Randomized controlled trial of behavioral treatment for comorbid obesity and depression in women: the Be Active Trial. Int. J. Obes. (Lond.) 37 (11), 1427–1434.
Pan, A., Keum, N., Okereke, O.I., et al., 2012. Bidirectional association between depression and metabolic syndrome: a systematic review and meta-analysis of epidemiological studies. Diabetes Care 35 (5), 1171–1180. Papakostas, G.I., Petersen, T., Iosifescu, D.V., et al., 2005. Obesity among outpatients with major depressive disorder. Int. J. Neuropsychopharmacol. 8 (1), 59–63. Pasquali, R., 2012. The hypothalamic–pituitary–adrenal axis and sex hormones in chronic stress and obesity: pathophysiological and clinical aspects. Ann. N. Y. Acad. Sci. 1264, 20–35. Pasquali, R., Vicennati, V., Cacciari, M., Pagotto, U., 2006. The hypothalamic–pituitary–adrenal axis activity in obesity and the metabolic syndrome. Ann. N. Y. Acad. Sci. 1083, 111–128. Patterson, Z.R., Khazall, R., Mackay, H., Anisman, H., Abizaid, A., 2013. Central ghrelin signaling mediates the metabolic response of C57BL/6 male mice to chronic social defeat stress. Endocrinology 154 (3), 1080–1091. Pawlby, S., Hay, D.F., Sharp, D., Waters, C.S., O’Keane, V., 2009. Antenatal depression predicts depression in adolescent offspring: prospective longitudinal community-based study. J. Affect. Disord. 113 (3), 236–243. Pearson, R.M., Evans, J., Kounali, D., et al., 2013. Maternal depression during pregnancy and the postnatal period: risks and possible mechanisms for offspring depression at age 18 years. JAMA Psychiatry 70 (12), 1312–1319. Pecoraro, N., Reyes, F., Gomez, F., Bhargava, A., Dallman, M.F., 2004. Chronic stress promotes palatable feeding, which reduces signs of stress: feedforward and feedback effects of chronic stress. Endocrinology 145 (8), 3754–3762. Perala, J., Suvisaari, J., Saarni, S.I., et al., 2007. Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch. Gen. Psychiatry 64 (1), 19–28. Pereira, D.S., de Queiroz, B.Z., Miranda, A.S., et al., 2013. Effects of physical exercise on plasma levels of brain-derived neurotrophic factor and depressive symptoms in elderly women – a randomized clinical trial. Arch. Phys. Med. Rehabil. 94 (8), 1443–1450. Pereira, T.S., Silva, A.A., Alves, M.T., et al., 2012. Perinatal and early life factors associated with symptoms of depression in Brazilian children. BMC Public Health 12, 605. Perlis, R.H., Ostacher, M.J., Patel, J.K., et al., 2006. Predictors of recurrence in bipolar disorder: primary outcomes from the Systematic Treatment Enhancement Program for Bipolar Disorder (STEP-BD). Am. J. Psychiatry 163 (2), 217–224. Pervanidou, P., Chrousos, G.P., 2012. Metabolic consequences of stress during childhood and adolescence. Metabolism 61 (5), 611–619. Petrovich, G.D., Holland, P.C., Gallagher, M., 2005. Amygdalar and prefrontal pathways to the lateral hypothalamus are activated by a learned cue that stimulates eating. J. Neurosci. 25 (36), 8295–8302. Pfaffenseller, B., Fries, G.R., Wollenhaupt-Aguiar, B., et al., 2013. Neurotrophins, inflammation and oxidative stress as illness activity biomarkers in bipolar disorder. Expert Rev. Neurother. 13 (7), 827–842. Phillips, M.L., Swartz, H.A., 2014. A critical appraisal of neuroimaging studies of bipolar disorder: toward a new conceptualization of underlying neural circuitry and a road map for future research. Am. J. Psychiatry. Pintana, H., Apaijai, N., Chattipakorn, N., Chattipakorn, S.C., 2013. DPP-4 inhibitors improve cognition and brain mitochondrial function of insulin-resistant rats. J. Endocrinol. 218 (1), 1–11. Pipatpiboon, N., Pintana, H., Pratchayasakul, W., Chattipakorn, N., Chattipakorn, S.C., 2013. DPP4-inhibitor improves neuronal insulin receptor function, brain mitochondrial function and cognitive function in rats with insulin resistance induced by high-fat diet consumption. Eur. J. Neurosci. 37 (5), 839–849. Porter, D.W., Kerr, B.D., Flatt, P.R., Holscher, C., Gault, V.A., 2010. Four weeks administration of Liraglutide improves memory and learning as well as glycaemic control in mice with high fat dietary-induced obesity and insulin resistance. Diabetes Obes. Metab. 12 (10), 891–899. Poston, L., 2012. Maternal obesity, gestational weight gain and diet as determinants of offspring long term health. Best Pract. Res. Clin. Endocrinol. Metab. 26 (5), 627–639. Primeau, V., Coderre, L., Karelis, A.D., et al., 2011. Characterizing the profile of obese patients who are metabolically healthy. Int. J. Obes. (Lond.) 35 (7), 971–981. Raisanen, S., Lehto, S.M., Nielsen, H.S., Gissler, M., Kramer, M.R., Heinonen, S., 2014. Risk factors for and perinatal outcomes of major depression during pregnancy: a population-based analysis during 2002–2010 in Finland. BMJ Open 4 (11), e004883. Raison, C.L., Rutherford, R.E., Woolwine, B.J., et al., 2013. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry 70 (1), 31–41. Reijmer, Y.D., Brundel, M., de Bresser, J., Kappelle, L.J., Leemans, A., Biessels, G.J., 2013a. Microstructural white matter abnormalities and cognitive functioning in type 2 diabetes: a diffusion tensor imaging study. Diabetes Care 36 (1), 137– 144. Reijmer, Y.D., Leemans, A., Brundel, M., Kappelle, L.J., Biessels, G.J., 2013b. Disruption of the cerebral white matter network is related to slowing of information processing speed in patients with type 2 diabetes. Diabetes. Rivera, M., Cohen-Woods, S., Kapur, K., et al., 2012. Depressive disorder moderates the effect of the FTO gene on body mass index. Mol. Psychiatry 17 (6), 604–611. Rotella, F., Mannucci, E., 2013a. Depression as a risk factor for diabetes: a metaanalysis of longitudinal studies. J. Clin. Psychiatry 74 (1), 31–37. Rotella, F., Mannucci, E., 2013b. Diabetes mellitus as a risk factor for depression. A meta-analysis of longitudinal studies. Diabetes Res. Clin. Pract. 99 (2), 98–104. Russo, S.J., Nestler, E.J., 2013. The brain reward circuitry in mood disorders. Nat. Rev. Neurosci. 14 (9), 609–625.
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104 Ruttle, P.L., Klein, M.H., Slattery, M.J., Kalin, N.H., Armstrong, J.M., Essex, M.J., 2014. Adolescent adrenocortical activity and adiposity: differences by sex and exposure to early maternal depression. Psychoneuroendocrinology 47, 68–77. Ruzickova, M., Slaney, C., Garnham, J., Alda, M., 2003. Clinical features of bipolar disorder with and without comorbid diabetes mellitus. Can. J. Psychiatry 48 (7), 458–461. Sahin, I., Alkan, A., Keskin, L., et al., 2008. Evaluation of in vivo cerebral metabolism on proton magnetic resonance spectroscopy in patients with impaired glucose tolerance and type 2 diabetes mellitus. J. Diabetes Complicat. 22 (4), 254–260. Samaan, Z., Anand, S.S., Zhang, X., et al., 2013. The protective effect of the obesityassociated rs9939609 A variant in fat mass- and obesity-associated gene on depression. Mol. Psychiatry 18 (12), 1281–1286. Sanchez-Villegas, A., Field, A.E., O’Reilly, E.J., et al., 2012. Perceived and actual obesity in childhood and adolescence and risk of adult depression. J. Epidemiol. Community Health. Sanhueza, C., Ryan, L., Foxcroft, D.R., 2013. Diet and the risk of unipolar depression in adults: systematic review of cohort studies. J. Hum. Nutr. Diet 26 (1), 56–70. Sassi, F., Devaux, M., Church, J., 2009. Education and Obesity in Four OECD Countries. OECD Health Working Paper No. 46. OECD Publishing, Paris, http://dx.doi.org/10.1787/5km4psmtn8zx-en. Schmoller, A., Hass, T., Strugovshchikova, O., et al., 2010. Evidence for a relationship between body mass and energy metabolism in the human brain. J. Cereb. Blood Flow Metab. 30 (7), 1403–1410. Schulkin, J., 2003. Allostasis: a neural behavioral perspective. Horm. Behav. 43 (1), 21–27 (discussion 28–30). Schulz, L.C., 2010. The Dutch Hunger Winter and the developmental origins of health and disease. Proc. Natl. Acad. Sci. U. S. A. 107 (39), 16757–16758. Schwartz, E., Mobbs, C.V., 2012. Hypothalamic BDNF and obesity: found in translation. Nat. Med. 18 (4), 496–497. Sepanjnia, K., Modabbernia, A., Ashrafi, M., Modabbernia, M.J., Akhondzadeh, S., 2012. Pioglitazone adjunctive therapy for moderate-to-severe major depressive disorder: randomized double-blind placebo-controlled trial. Neuropsychopharmacology 37 (9), 2093–2100. Sharma, A.N., Elased, K.M., Garrett, T.L., Lucot, J.B., 2010. Neurobehavioral deficits in db/db diabetic mice. Physiol. Behav. 101 (3), 381–388. Sharma, S., Fulton, S., 2013. Diet-induced obesity promotes depressive-like behaviour that is associated with neural adaptations in brain reward circuitry. Int. J. Obes. (Lond.) 37 (3), 382–389. Shugart, Y.Y., Chen, L., Day, I.N., et al., 2009. Two British women studies replicated the association between the Val66Met polymorphism in the brain-derived neurotrophic factor (BDNF) and BMI. Eur. J. Hum. Genet. 17 (8), 1050–1055. Sicras, A., Rejas, J., Navarro, R., Serrat, J., Blanca, M., 2008. Metabolic syndrome in bipolar disorder: a cross-sectional assessment of a Health Management Organization database. Bipolar Disord. 10 (5), 607–616. Siervo, M., Arnold, R., Wells, J.C., et al., 2011. Intentional weight loss in overweight and obese individuals and cognitive function: a systematic review and metaanalysis. Obes. Rev. 12 (11), 968–983. Silvestri, C., Di Marzo, V., 2012. Second generation CB1 receptor blockers and other inhibitors of peripheral endocannabinoid overactivity and the rationale of their use against metabolic disorders. Expert Opin. Investig. Drugs 21 (9), 1309–1322. Simon, G.E., Ludman, E.J., Linde, J.A., et al., 2008. Association between obesity and depression in middle-aged women. Gen. Hosp. Psychiatry 30 (1), 32–39. Simon, G.E., Rohde, P., Ludman, E.J., et al., 2010. Association between change in depression and change in weight among women enrolled in weight loss treatment. Gen. Hosp. Psychiatry 32 (6), 583–589. Skjelstad, D.V., Malt, U.F., Holte, A., 2010. Symptoms and signs of the initial prodrome of bipolar disorder: a systematic review. J. Affect. Disord. 126 (1–2), 1–13. Smoller, J.W., Finn, C.T., 2003. Family, twin, and adoption studies of bipolar disorder. Am. J. Med. Genet. C: Semin. Med. Genet. 123C (1), 48–58. Snyder, H.R., 2013. Major depressive disorder is associated with broad impairments on neuropsychological measures of executive function: a meta-analysis and review. Psychol. Bull. 139 (1), 81–132. Sozeri-Varma, G., Kalkan-Oguzhanoglu, N., Efe, M., Kiroglu, Y., Duman, T., 2013. Neurochemical metabolites in prefrontal cortex in patients with mild/moderate levels in first-episode depression. Neuropsychiatr. Dis. Treat. 9, 1053–1059. Stefan, N., Kantartzis, K., Machann, J., et al., 2008. Identification and characterization of metabolically benign obesity in humans. Arch. Intern. Med. 168 (15), 1609–1616. Steptoe, A., Kunz-Ebrecht, S.R., Owen, N., 2003. Lack of association between depressive symptoms and markers of immune and vascular inflammation in middle-aged men and women. Psychol. Med. 33 (4), 667–674. Sterling, P., 2012. Allostasis: a model of predictive regulation. Physiol. Behav. 106 (1), 5–15. Stetler, C., Miller, G.E., 2011. Depression and hypothalamic–pituitary–adrenal activation: a quantitative summary of four decades of research. Psychosomal Med. 73 (2), 114–126. Stork, C., Renshaw, P.F., 2005. Mitochondrial dysfunction in bipolar disorder: evidence from magnetic resonance spectroscopy research. Mol. Psychiatry 10 (10), 900–919. Strachan, M.W., Deary, I.J., Ewing, F.M., Frier, B.M., 1997. Is type II diabetes associated with an increased risk of cognitive dysfunction? A critical review of published studies. Diabetes Care 20 (3), 438–445. Strakowski, S.M., Adler, C.M., Almeida, J., et al., 2012. The functional neuroanatomy of bipolar disorder: a consensus model. Bipolar Disord. 14 (4), 313–325.
103
Stranahan, A.M., Norman, E.D., Lee, K., et al., 2008. Diet-induced insulin resistance impairs hippocampal synaptic plasticity and cognition in middle-aged rats. Hippocampus 18 (11), 1085–1088. Strekalova, T., Couch, Y., Kholod, N., et al., 2011. Update in the methodology of the chronic stress paradigm: internal control matters. Behav. Brain Funct. 7, 9. Stunkard, A.J., Foch, T.T., Hrubec, Z., 1986. A twin study of human obesity. JAMA 256 (1), 51–54. Sullivan, P.F., Neale, M.C., Kendler, K.S., 2000. Genetic epidemiology of major depression: review and meta-analysis. Am. J. Psychiatry 157 (10), 1552–1562. Sussulini, A., Prando, A., Maretto, D.A., et al., 2009. Metabolic profiling of human blood serum from treated patients with bipolar disorder employing 1 H NMR spectroscopy and chemometrics. Anal. Chem. 81 (23), 9755–9763. Swift, D.L., Johannsen, N.M., Lavie, C.J., Earnest, C.P., Church, T.S., 2014. The role of exercise and physical activity in weight loss and maintenance. Prog. Cardiovasc. Dis. 56 (4), 441–447. Tamashiro, K.L., Sakai, R.R., Shively, C.A., Karatsoreos, I.N., Reagan, L.P., 2011. Chronic stress, metabolism, and metabolic syndrome. Stress 14 (5), 468–474. Tanaka, T., Ngwa, J.S., van Rooij, F.J., et al., 2013. Genome-wide meta-analysis of observational studies shows common genetic variants associated with macronutrient intake. Am. J. Clin. Nutr. 97 (6), 1395–1402. Taylor, V.H., MacQueen, G.M., 2007. Cognitive dysfunction associated with metabolic syndrome. Obes. Rev. 8 (5), 409–418. Teh, A.L., Pan, H., Chen, L., et al., 2014. The effect of genotype and in utero environment on interindividual variation in neonate DNA methylomes. Genome Res. 24 (7), 1064–1074. Thomas, G., Khunti, K., Curcin, V., et al., 2014. Obesity paradox in people newly diagnosed with type 2 diabetes with and without prior cardiovascular disease. Diabetes Obes. Metab. 16 (4), 317–325. Thrush, A.B., Dent, R., McPherson, R., Harper, M.E., 2013. Implications of mitochondrial uncoupling in skeletal muscle in the development and treatment of obesity. FEBS J. 280 (20), 5015–5029. Tobias, D.K., Pan, A., Jackson, C.L., et al., 2014. Body-mass index and mortality among adults with incident type 2 diabetes. N. Engl. J. Med. 370 (3), 233–244. Toups, M.S., Myers, A.K., Wisniewski, S.R., et al., 2013. Relationship between obesity and depression: characteristics and treatment outcomes with antidepressant medication. Psychosomal Med. 75 (9), 863–872. Tremblay, A., Chaput, J.P., 2012. Obesity: the allostatic load of weight loss dieting. Physiol. Behav. 106 (1), 16–21. Tryon, M.S., DeCant, R., Laugero, K.D., 2013. Having your cake and eating it too: a habit of comfort food may link chronic social stress exposure and acute stress-induced cortisol hyporesponsiveness. Physiol. Behav. 114–115, 32–37. Uher, R., Mors, O., Hauser, J., et al., 2009. Body weight as a predictor of antidepressant efficacy in the GENDEP project. J. Affect. Disord. 118 (1–3), 147–154. Unger, T.J., Calderon, G.A., Bradley, L.C., Sena-Esteves, M., Rios, M., 2007. Selective deletion of Bdnf in the ventromedial and dorsomedial hypothalamus of adult mice results in hyperphagic behavior and obesity. J. Neurosci. 27 (52), 14265–14274. van de Giessen, E., Celik, F., Schweitzer, D.H., van den Brink, W., Booij, J., 2014. Dopamine D2/3 receptor availability and amphetamine-induced dopamine release in obesity. J. Psychopharmacol. 28 (9), 866–873. van Duinkerken, E., Schoonheim, M.M., Ijzerman, R.G., et al., 2012a. Diffusion tensor imaging in type 1 diabetes: decreased white matter integrity relates to cognitive functions. Diabetologia 55 (4), 1218–1220. van Duinkerken, E., Schoonheim, M.M., Sanz-Arigita, E.J., et al., 2012b. Resting-state brain networks in type 1 diabetic patients with and without microangiopathy and their relation to cognitive functions and disease variables. Diabetes 61 (7), 1814–1820. van Loo, H.M., de Jonge, P., Romeijn, J.W., Kessler, R.C., Schoevers, R.A., 2012. Datadriven subtypes of major depressive disorder: a systematic review. BMC Med. 10, 156. Vancampfort, D., Correll, C.U., Probst, M., et al., 2013a. A review of physical activity correlates in patients with bipolar disorder. J. Affect. Disord. 145 (3), 285–291. Vancampfort, D., Vansteelandt, K., Correll, C.U., et al., 2013b. Metabolic syndrome and metabolic abnormalities in bipolar disorder: a meta-analysis of prevalence rates and moderators. Am. J. Psychiatry 170 (3), 265–274. Vannucchi, G., Toni, C., Maremmani, I., Perugi, G., 2014. Does obesity predict bipolarity in major depressive patients? J. Affect. Disord. 155, 118–122. Vargas, C., Lopez-Jaramillo, C., Vieta, E., 2013. A systematic literature review of resting state network-functional MRI in bipolar disorder. J. Affect. Disord. 150 (3), 727–735. Vederine, F.E., Wessa, M., Leboyer, M., Houenou, J., 2011. A meta-analysis of whole-brain diffusion tensor imaging studies in bipolar disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry 35 (8), 1820–1826. Veen, G., Giltay, E.J., DeRijk, R.H., van Vliet, I.M., van Pelt, J., Zitman, F.G., 2009. Salivary cortisol, serum lipids, and adiposity in patients with depressive and anxiety disorders. Metabolism 58 (6), 821–827. Vernochet, C., Kahn, C.R., 2012. Mitochondria, obesity and aging. Aging (Albany, NY) 4 (12), 859–860. Vogelzangs, N., Beekman, A.T., Boelhouwer, I.G., et al., 2011. Metabolic depression: a chronic depressive subtype? Findings from the InCHIANTI study of older persons. J. Clin. Psychiatry 72 (5), 598–604. Vogelzangs, N., Duivis, H.E., Beekman, A.T., et al., 2012. Association of depressive disorders, depression characteristics and antidepressant medication with inflammation. Transl. Psychiatry 2, e79.
104
R.B. Mansur et al. / Neuroscience and Biobehavioral Reviews 52 (2015) 89–104
Vogelzangs, N., Kritchevsky, S.B., Beekman, A.T., et al., 2010. Obesity and onset of significant depressive symptoms: results from a prospective community-based cohort study of older men and women. J. Clin. Psychiatry 71 (4), 391–399. Volkow, N.D., Wang, G.J., Baler, R.D., 2011. Reward, dopamine and the control of food intake: implications for obesity. Trends Cogn. Sci. 15 (1), 37–46. Volkow, N.D., Wang, G.J., Fowler, J.S., Telang, F., 2008. Overlapping neuronal circuits in addiction and obesity: evidence of systems pathology. Philos. Trans. R. Soc. Lond. B: Biol. Sci. 363 (1507), 3191–3200. Vos, T., Flaxman, A.D., Naghavi, M., et al., 2012. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380 (9859), 2163–2196. Vreeburg, S.A., Hoogendijk, W.J., van Pelt, J., et al., 2009. Major depressive disorder and hypothalamic–pituitary–adrenal axis activity: results from a large cohort study. Arch. Gen. Psychiatry 66 (6), 617–626. Wang-Sattler, R., Yu, Z., Herder, C., et al., 2012. Novel biomarkers for pre-diabetes identified by metabolomics. Mol. Syst. Biol. 8, 615. Wang, L., Anderson, J.L., Dalton III, W.T., Wu, T., Liu, X., Zheng, S., 2013. Maternal depressive symptoms and the risk of overweight in their children. Matern. Child Health J. 17 (5), 940–948. Wang, T.J., Larson, M.G., Vasan, R.S., et al., 2011. Metabolite profiles and the risk of developing diabetes. Nat. Med. 17 (4), 448–453. Watson, S., Gallagher, P., Dougall, D., et al., 2013. Childhood trauma in bipolar disorder. Aust. N. Z. J. Psychiatry. Werrij, M.Q., Mulkens, S., Hospers, H.J., Jansen, A., 2006. Overweight and obesity: the significance of a depressed mood. Patient Educ. Couns. 62 (1), 126–131. Whiteford, H.A., Harris, M.G., McKeon, G., et al., 2013. Estimating remission from untreated major depression: a systematic review and meta-analysis. Psychol. Med. 43 (8), 1569–1585. Whitmer, R.A., Gunderson, E.P., Barrett-Connor, E., Quesenberry Jr., C.P., Yaffe, K., 2005. Obesity in middle age and future risk of dementia: a 27 year longitudinal population based study. BMJ 330 (7504), 1360. Wildman, R.P., Muntner, P., Reynolds, K., et al., 2008. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999–2004). Arch. Intern. Med. 168 (15), 1617– 1624. Wilhelm, C.J., Choi, D., Huckans, M., Manthe, L., Loftis, J.M., 2013. Adipocytokine signaling is altered in Flinders sensitive line rats, and adiponectin correlates in humans with some symptoms of depression. Pharmacol. Biochem. Behav. 103 (3), 643–651. Winham, S.J., Biernacka, J.M., 2013. Gene–environment interactions in genome-wide association studies: current approaches and new directions. J. Child Psychol. Psychiatry 54 (10), 1120–1134. Wojcik, W., Lee, W., Colman, I., Hardy, R., Hotopf, M., 2013. Foetal origins of depression? A systematic review and meta-analysis of low birth weight and later depression. Psychol. Med. 43 (1), 1–12.
Wong, M.L., Kling, M.A., Munson, P.J., et al., 2000. Pronounced and sustained central hypernoradrenergic function in major depression with melancholic features: relation to hypercortisolism and corticotropin-releasing hormone. Proc. Natl. Acad. Sci. U. S. A. 97 (1), 325–330. Woo, J., Shin, K.O., Park, S.Y., Jang, K.S., Kang, S., 2013. Effects of exercise and diet change on cognition function and synaptic plasticity in high fat diet induced obese rats. Lipids Health Dis. 12, 144. Wu, C.S., Nohr, E.A., Bech, B.H., Vestergaard, M., Olsen, J., 2012. Long-term health outcomes in children born to mothers with diabetes: a population-based cohort study. PLoS ONE 7 (5), e36727. Xia, Q., Grant, S.F., 2013. The genetics of human obesity. Ann. N. Y. Acad. Sci. 1281, 178–190. Xu, B., Goulding, E.H., Zang, K., et al., 2003. Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nat. Neurosci. 6 (7), 736–742. Xu, J., Dydak, U., Harezlak, J., et al., 2013. Neurochemical abnormalities in unmedicated bipolar depression and mania: a 2D (1)H MRS investigation. Psychiatry Res. 213 (3), 235–241. Xu, W.L., Atti, A.R., Gatz, M., Pedersen, N.L., Johansson, B., Fratiglioni, L., 2011. Midlife overweight and obesity increase late-life dementia risk: a population-based twin study. Neurology 76 (18), 1568–1574. Yang, N., Ginsburg, G.S., Simmons, L.A., 2013. Personalized medicine in women’s obesity prevention and treatment: implications for research, policy and practice. Obes. Rev. 14 (2), 145–161. Yanovski, S.Z., Yanovski, J.A., 2014. Long-term drug treatment for obesity: a systematic and clinical review. JAMA 311 (1), 74–86. Yim, C.Y., Soczynska, J.K., Kennedy, S.H., Woldeyohannes, H.O., Brietzke, E., McIntyre, R.S., 2012. The effect of overweight/obesity on cognitive function in euthymic individuals with bipolar disorder. Eur. Psychiatry 27 (3), 223–228. Zagrebelsky, M., Korte, M., 2014. Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology 76 (Pt C), 628–638. Zeman, M., Jirak, R., Jachymova, M., Vecka, M., Tvrzicka, E., Zak, A., 2009. Leptin, adiponectin, leptin to adiponectin ratio and insulin resistance in depressive women. Neuroendocrinol Lett. 30 (3), 387–395. Zhao, G., Ford, E.S., Dhingra, S., Li, C., Strine, T.W., Mokdad, A.H., 2009. Depression and anxiety among US adults: associations with body mass index. Int. J. Obes. (Lond.) 33 (2), 257–266. Zheng, P., Wang, Y., Chen, L., et al., 2013. Identification and validation of urinary metabolite biomarkers for major depressive disorder. Mol. Cell Proteomics 12 (1), 207–214. Zhou, J.X., Li, H.C., Bai, X.J., et al., 2013. Functional Val66Met polymorphism of brainderived neurotrophic factor in type 2 diabetes with depression in Han Chinese subjects. Behav. Brain Funct. 9, 34. Zupancic, M.L., Mahajan, A., 2011. Leptin as a neuroactive agent. Psychosomal Med. 73 (5), 407–414.